Download Translational models of coronary artery disease, myocardial

Document related concepts

Cardiac contractility modulation wikipedia , lookup

Heart failure wikipedia , lookup

Electrocardiography wikipedia , lookup

Saturated fat and cardiovascular disease wikipedia , lookup

Remote ischemic conditioning wikipedia , lookup

Echocardiography wikipedia , lookup

Cardiovascular disease wikipedia , lookup

Antihypertensive drug wikipedia , lookup

Arrhythmogenic right ventricular dysplasia wikipedia , lookup

Quantium Medical Cardiac Output wikipedia , lookup

Cardiac surgery wikipedia , lookup

Drug-eluting stent wikipedia , lookup

Angina wikipedia , lookup

Dextro-Transposition of the great arteries wikipedia , lookup

History of invasive and interventional cardiology wikipedia , lookup

Coronary artery disease wikipedia , lookup

Management of acute coronary syndrome wikipedia , lookup

Transcript
TRANSLATIONAL MODELS OF
CORONARY ARTERY DISEASE,
MYOCARDIAL INFARCTION
AND HEART FAILURE
Development, Validation and in vivo Imaging Studies
Using Positron Emission Tomography
Miikka Tarkia
TURUN YLIOPISTON JULKAISUJA – ANNALES UNIVERSITATIS TURKUENSIS
Sarja - ser. D osa - tom. 1175 | Medica - Odontologica | Turku 2015
University of Turku
Faculty of Medicine
Institute of Clinical Medicine
Department of Clinical Physiology and Nuclear Medicine and Turku PET Centre
Drug Research Doctoral Programme at University of Turku
Supervised by
Professor Anne Roivainen, PhD
Turku PET Centre and Turku Centre for Disease Modelling
University of Turku
Turku, Finland
Associate Professor Antti Saraste, MD, PhD
Turku PET Centre and Heart Center
University of Turku and Turku University Hospital
Turku, Finland
Reviewed by
Adjunct Professor Esko Kankuri, MD, PhD
Department of Pharmacology
University of Helsinki
Helsinki, Finland
Adjunct Professor Kirsi Timonen, MD, PhD
Department of Clinical Physiology and Nuclear Medicine
Central Hospital of Central Finland and University of Eastern Finland
Kuopio, Finland
Opponent
Professor Johanna Kuusisto, MD, PhD
Department of Medicine
University of Eastern Finland
Kuopio, Finland
The originality of this thesis has been checked in accordance with the University of Turku quality
assurance system using the Turnitin OriginalityCheck service.
ISBN 978-951-29-6129-0 (PRINT)
ISBN 978-951-29-6130-6 (PDF)
ISSN 0355-9483
Painosalama Oy - Turku, Finland 2015
To Anna and Maria
4
Abstract
ABSTRACT
Miikka Tarkia
TRANSLATIONAL MODELS OF CORONARY ARTERY DISEASE,
MYOCARDIAL INFARCTION AND HEART FAILURE
Development, validation and in vivo imaging studies using positron emission tomography
Department of Clinical Physiology and Nuclear Medicine and Turku PET Centre,
University of Turku, Turku, Finland
Coronary artery disease is an atherosclerotic disease, which leads to narrowing of coronary arteries,
deteriorated myocardial blood flow and myocardial ischaemia. In acute myocardial infarction, a
prolonged period of myocardial ischaemia leads to myocardial necrosis. Necrotic myocardium
is replaced with scar tissue. Myocardial infarction results in various changes in cardiac structure
and function over time that results in “adverse remodelling”. This remodelling may result in a
progressive worsening of cardiac function and development of chronic heart failure.
In this thesis, we developed and validated three different large animal models of coronary
artery disease, myocardial ischaemia and infarction for translational studies. In the first study
the coronary artery disease model had both induced diabetes and hypercholesterolemia. In
the second study myocardial ischaemia and infarction were caused by a surgical method
and in the third study by catheterisation. For model characterisation, we used non-invasive
positron emission tomography (PET) methods for measurement of myocardial perfusion,
oxidative metabolism and glucose utilisation. Additionally, cardiac function was measured
by echocardiography and computed tomography. To study the metabolic changes that occur
during atherosclerosis, a hypercholesterolemic and diabetic model was used with [18F]
fluorodeoxyglucose ([18F]FDG) PET-imaging technology. Coronary occlusion models were
used to evaluate metabolic and structural changes in the heart and the cardioprotective effects
of levosimendan during post-infarction cardiac remodelling. Large animal models were used
in testing of novel radiopharmaceuticals for myocardial perfusion imaging.
In the coronary artery disease model, we observed atherosclerotic lesions that were associated
with focally increased [18F]FDG uptake. In heart failure models, chronic myocardial infarction
led to the worsening of systolic function, cardiac remodelling and decreased efficiency of
cardiac pumping function. Levosimendan therapy reduced post-infarction myocardial infarct
size and improved cardiac function. The novel 68Ga-labeled radiopharmaceuticals tested in
this study were not successful for the determination of myocardial blood flow.
In conclusion, diabetes and hypercholesterolemia lead to the development of early phase
atherosclerotic lesions. Coronary artery occlusion produced considerable myocardial
ischaemia and later infarction following myocardial remodelling. The experimental models
evaluated in these studies will enable further studies concerning disease mechanisms, new
radiopharmaceuticals and interventions in coronary artery disease and heart failure.
Keywords: coronary artery disease, myocardial ischaemia, myocardial infarction, heart
failure, positron emission tomography (PET)
Tiivistelmä5
TIIVISTELMÄ
Miikka Tarkia
SEPELVALTIMOTAUDIN, SYDÄNINFARKTIN JA SYDÄMEN
VAJAATOIMINNAN TRANSLATIONAALISET MALLIT
Kehitys, validointi ja in vivo kuvantaminen positroniemissiotomografialla
Kliininen fysiologia ja isotooppilääketiede ja Valtakunnallinen PET-keskus, Turun
yliopisto, Turku, Suomi
Sepelvaltimotaudissa sepelvaltimot ahtautuvat ateroskleroosin vuoksi, mikä johtaa sydänlihaksen verenvirtauksen heikkenemiseen ja sydänlihasiskemiaan. Akuutissa sydäninfarktissa
pitkittynyt iskemia johtaa sydänlihaskuolioon eli nekroosiin, joka korvautuu arpikudoksella.
Sydäninfarktin seurauksena sydämen rakenteessa ja toiminnassa tapahtuu muutoksia, mikä
johtaa niin kutsuttuun haitalliseen sydänlihaksen uudelleenmuovautumiseen ja lopulta sydämen toiminnan heikkenemiseen ja kroonisen vajaatoiminnan kehittymiseen.
Tässä väitöskirjatyössä kehitettiin ja validoitiin kolme erilaista sepelvaltimotaudin, sydänlihasiskemian ja infarktin suureläinmallia translationaalisia tutkimuksia varten. Ensimmäisessä osatyössä sepelvaltimotaudin mallissa oli yhdistetty diabetes ja hyperkolesterolemia.
Toisessa osatyössä sydänlihasiskemia ja infarkti oli aiheutettu kirurgisesti ja kolmannessa
osatyössä katetrisaatiolla. Mallien karakterisoinnissa käytettiin kajoamattomia positroni­
emissiotomografiaan (PET) perustuvia kuvantamismenetelmiä, joilla mitattiin sydänlihaksen verenvirtausta, hapenkulutusta ja glukoosinkäyttöä. Lisäksi sydämen pumppausominaisuuksia mitattiin ultraäänikardiografialla ja tietokonetomografialla. Hyperkolesterolemia ja
diabetesmallilla tutkittiin ateroskleroottisten tautimuutosten kuvantamista [18F]fluorodeoksi­
glukoosi ([18F]FDG) PET-merkkiaineella. Sepelvaltimon ahtauman malleilla tutkittiin sydämen metabolisia ja rakenteellisia muutoksia ja levosimendaanin sydäntä suojaavaa vaikutusta.
Tutkimme myös uusia radiolääkeaineita sydänlihaksen verenvirtauksen määrittämiseen.
Sepelvaltimotaudin mallissa havaitsimme ateroskleroottisia muutoksia, jotka olivat yhteydessä lisääntyneeseen [18F]FDG kertymään. Sydämen vajaatoiminnan malleissa krooninen
sydäninfarkti johti systolisen funktion huononemiseen, haitalliseen sydänlihaksen uudelleenmuovautumiseen ja alentuneeseen sydänlihaksen hyötysuhteeseen. Infarktinjälkeinen
hoito levosimendaanilla johti infarktialueen rajoittumiseen ja sydämen pumppausominaisuuksien parantumiseen. Uudet testatut 68Ga-leimatut radiolääkeaineet eivät tässä tutkimuksessa soveltuneet sydänlihaksen verenvirtauksen määrittämiseen.
Johtopäätöksenä, diabetes ja hyperkolesterolemia johtavat varhaisen vaiheen ateroskleroottisiin muutoksiin. Sepelvaltimon tukkeuma kehitti merkittävän sydänlihasiskemian ja myöhemmän infarktin johtaen sydänlihaksen uudelleenmuovautumiseen. Testattujen koemallien
myötä meillä on mahdollisuus tutkia lisää sepelvaltimotaudin ja sydämen vajaatoiminnan
tautimekanismeja, uusia radiolääkeaineita ja hoitomuotoja.
Avainsanat: sepelvaltimotauti, sydänlihasiskemia, sydäninfarkti, sydämen vajaatoiminta,
positroniemissiotomografia (PET)
6
Table of Contents
TABLE OF CONTENTS
ABSTRACT.....................................................................................................................4
TIIVISTELMÄ...............................................................................................................5
ABBREVIATIONS.........................................................................................................8
LIST OF ORIGINAL PUBLICATIONS....................................................................10
1.INTRODUCTION................................................................................................... 11
2. REVIEW OF THE LITERATURE........................................................................13
2.1 Coronary artery disease......................................................................................13
2.1.1 Atherosclerotic inflammation and plaque formation................................13
2.1.2 Coronary artery stenosis and plaque rupture............................................14
2.2 Ischaemic cardiomyopathy.................................................................................14
2.2.1 Myocardial ischaemia..............................................................................14
2.2.2 Ischaemic preconditioning.......................................................................15
2.2.3 Myocardial infarction...............................................................................16
2.2.4 Cardiac remodelling.................................................................................16
2.2.5 Heart failure..............................................................................................17
2.3 Experimental models of coronary artery disease................................................18
2.3.1 Large animal models of atherosclerosis...................................................19
2.4 Experimental models of ischaemic cardiomyopathy..........................................21
2.4.1 Large animal models of chronic myocardial ischaemia...........................22
2.4.2 Large animal models of acute ischaemia-reperfusion injury and infarction.. 24
2.4.3 Large animal models of chronic myocardial infarction,
remodelling and heart failure...................................................................25
2.5 Imaging of coronary artery disease and ischaemic cardiomyopathy..................28
2.5.1 Invasive coronary artery imaging.............................................................28
2.5.2 Molecular imaging of atherosclerotic inflammation................................28
2.5.3 Left ventricular function and structure.....................................................30
2.5.4 Myocardial perfusion...............................................................................30
2.5.5 Myocardial infarction and viability..........................................................31
2.5.6 PET imaging of myocardial metabolism..................................................32
2.6 Positive inotropic therapy in heart failure..........................................................32
3. AIMS OF THE STUDY...........................................................................................34
4. MATERIALS AND METHODS.............................................................................35
4.1 Coronary artery disease model (I)......................................................................35
4.1.1 Experimental protocol..............................................................................35
4.1.2 In vivo PET imaging of atherosclerotic inflammation.............................35
4.1.3 Ex vivo studies.........................................................................................35
4.2 Surgical and percutaneous myocardial infarction model (II, III).......................36
4.2.1 Experimental protocol..............................................................................36
4.2.2 PET imaging of myocardial perfusion and viability................................37
Table of Contents7
4.2.3 Left ventricular size and function (II)......................................................37
4.2.4 Myocardial oxidative metabolism and efficiency (II)..............................38
4.2.5 Tissue samples and histology...................................................................38
4.3 New myocardial perfusion tracers (IV)..............................................................38
4.3.1 Study design.............................................................................................38
4.3.2 PET imaging and kinetic modelling of [68Ga] ligands.............................39
4.3.3 Organ distribution....................................................................................39
4.3.4 In vitro binding to serum proteins............................................................39
4.4 Chronic levosimendan therapy for heart failure (V)..........................................39
4.4.1 Study design.............................................................................................39
4.4.2 Effects of chronic levosimendan therapy on MI size, LV function
and remodelling........................................................................................40
4.5 Statistical analyses..............................................................................................40
5.RESULTS.................................................................................................................41
5.1 Atherosclerotic plaque inflammation (I)............................................................41
5.1.1 Ex vivo studies.........................................................................................41
5.1.2 In vivo PET imaging................................................................................41
5.2 Myocardial infarction and remodelling (II, III)..................................................42
5.2.1 Myocardial perfusion and viability..........................................................42
5.2.2 Left ventricular function (II)....................................................................43
5.2.3 Myocardial oxidative metabolism and efficiency (II)..............................43
5.2.4 Tissue samples and histology (II).............................................................43
5.3 Evaluation of new perfusion tracers (IV)...........................................................43
5.3.1 PET imaging and kinetic modelling of [68Ga] ligands.............................43
5.3.2 Organ distribution....................................................................................44
5.3.3 In vitro binding to serum proteins............................................................44
5.4 Evaluation of chronic levosimendan intervention for heart failure (V).............44
5.4.1 Effects of chronic levosimendan therapy on MI size, LV function
and remodelling........................................................................................44
6.DISCUSSION...........................................................................................................46
6.1 PET imaging of early atherosclerotic lesions.....................................................46
6.2 Validation of a surgically induced model of myocardial infarction...................47
6.3 Validation of percutaneously-induced model of myocardial ischaemia and
infarction.............................................................................................................48
6.4 PET imaging and kinetic modelling of [68Ga] ligands........................................48
6.5 Chronic levosimendan intervention for heart failure..........................................49
6.6 Critical evaluation of the results.........................................................................49
6.7 Future aspects.....................................................................................................50
7. SUMMARY AND CONCLUSIONS.......................................................................52
9.REFERENCES........................................................................................................55
10.ORIGINAL PUBLICATIONS................................................................................73
8
Abbreviations
ABBREVIATIONS
AC
Adenylyl cyclase
ADP
Adenosine diphosphate
Alpha-V beta-3 integrin
αvβ3
AHA
American Heart Association
Akt
Protein kinase B
AMP
Adenosine monophosphate
ATP
Adenosine triphosphate
18
2-[18F]-Fluoro-2-deoxy-D-glucose
[ F]FDG
BAPEN
N,N′-bis(3-aminopropyl)ethylenediamine
BAPDMEN N,N′-bis(3-aminopropyl)-dimethylenediamine
CFR
Coronary flow reserve
COX-2
Cyclooxygenase-2
CT
Computed tomography
CVR
Coronary vascular resistance
DOTATATE 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid octreotate
ECGElectrocardiography
EDV
End-diastolic volume
EF5
2-(2-Nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide
EF
Ejection fraction
ESV
End-systolic volume
FFA
Free fatty acids
FDA
United States Food and Drug Administration
FMISO
Fluoromisonidazole
FR
Folate receptor
Stimulatory G-protein
Gs
GSK-3β
Glycogen synthase kinase-3β
HF
Heart failure
HO-1
Heme oxygenase-1
IL
Interleukin
iNOS
Inducible Nitric-Oxide Synthase
IVUS
Intravascular ultrasound
JAK
Janus kinase
Adenosine triphosphate-sensitive potassium channel
KATP
LAD
Left anterior descending coronary artery
LCX
Left circumflex artery
LDL
Low density lipoprotein
LV
Left ventricle
MAPK
MBF
MCP-1
M-CSF
MFR
MI
MMP
MnSOD
MPI
MR
MRI
NF-κB
NO
OCT
PCSK9
PET
PI3K
PK11195
PKA
PKC
PTI
PTF
RCA
RGD
ROI
RPP
SR
SPECT
STAT
SUV
TGF-β1 TK
TLR
TNFα
TSPO
TTC
VCAM-1
VOI
WMI
Abbreviations9
Mitogen-activated protein kinase
Myocardial blood flow
Monocyte chemotactic protein 1
Macrophage-colony stimulating factor
Myocardial flow reserve
Myocardial infarction
Matrix metalloproteinase
Manganese superoxide dismutase
Myocardial perfusion imaging
Mannose receptor
Magnetic resonance imaging
Nuclear factor kappa-B
Nitric oxide
Optical coherence tomography
Proprotein convertase subtilisin/kexin type 9
Positron emission tomography
Phosphatidylinositol-3-kinase
N-butan-2-yl-1-(2-chlorophenyl)-N-methylisoquinoline-3-carboxamide
Protein kinase A
Protein kinase C
Perfusable tissue index
Perfusable tissue fraction
Right coronary artery
Tripeptide arginylglycylaspartic acid
Region of interest
Rate-pressure product
Scavenger receptor
Single-photon emission computed tomography
Signal transducers and activators of transcription
Standardised uptake value
Transforming growth factor-β1
Tyrosine kinase
Toll-like receptor
Tumour necrosis factor alpha
Translocator protein
2,3,5-triphenyl-tetrazolium chloride
Vascular cell adhesion molecule 1
Volume of interest
Work-metabolic index
10
List of Original Publications
LIST OF ORIGINAL PUBLICATIONS
This thesis is based on the following original publications
I.
Tarkia M, Saraste A, Stark C, Vähäsilta T, Savunen T, Strandberg M, Saunavaara
V, Tolvanen T, Teuho J, Teräs M, Metsälä O, Rinne P, Heinonen I, Savisto N,
Pietilä M, Saukko P, Roivainen A, Knuuti J. [18F]FDG accumulation in early
coronary atherosclerotic lesions in pigs. Manuscript submitted for publication.
II.
Tarkia M, Stark C, Haavisto M, Kentala R, Vähäsilta T, Savunen T, Strandberg M,
Hynninen V-V, Saunavaara V, Tolvanen T, Teräs M, Rokka J, Pietilä M, Saukko
P, Roivainen A, Saraste A, Knuuti J. Cardiac remodeling in a new pig model of
chronic heart failure: assessment of left ventricular functional, metabolic and
structural changes using PET, CT and echocardiography. Journal of Nuclear
Cardiology, published online 20 February 2015.
III.
Rissanen TT, Nurro J, Halonen PJ, Tarkia M, Saraste A, Rannankari M,
Honkonen K, Pietilä M, Leppänen O, Kuivanen A, Knuuti J, Ylä-Herttuala S.
The bottleneck stent model for chronic myocardial ischemia and heart failure
in pigs. American Journal of Physiology - Heart and Circulatory Physiology.
2013;305(9):H1297-H1308.
IV.
Tarkia M, Saraste A, Saanijoki T, Oikonen V, Vähäsilta T, Strandberg M, Stark C,
Tolvanen T, Teräs M, Savunen T, Green MA, Knuuti J, Roivainen A. Evaluation
of 68Ga-labeled tracers for PET imaging of myocardial perfusion in pigs. Nuclear
Medicine and Biology. 2012;39(5):715-723.
V.
Tarkia M, Stark C, Haavisto M, Kentala R, Vähäsilta T, Savunen T, Strandberg
M, Saunavaara V, Tolvanen T, Teräs M, Pietilä M, Nyman L, Duvall E, Saukko P,
Levijoki J, Roivainen A, Saraste A, Knuuti J. Effect of levosimendan therapy on
myocardial infarct size and left ventricle function after acute coronary occlusion.
A study with experimental pig model of post-infarction heart failure. Manuscript
submitted for publication.
The original communications have been reproduced with the permissions of the copyright
holders.
1.
Introduction11
INTRODUCTION
Ischaemic heart disease is the leading cause of death worldwide. It is caused by coronary
artery disease (CAD), which is a slowly progressing disease in which inflammatory
processes together with cholesterol accumulation lead to the narrowing of the coronary
artery. Narrowing of a coronary artery causes insufficient myocardial blood flow that
leads to myocardial ischaemia, which is defined as reduced myocardial oxygen supply
and metabolic changes due to decreased blood flow. Severely disturbed coronary blood
flow or rupture of an atherosclerotic plaque resulting in intraluminal thrombosis can
cause prolonged periods of ischaemia resulting in myocardial necrosis, i.e., acute
myocardial infarction (MI). MI is followed by adverse cardiac remodelling including
changes in size, shape, structure and physiology of the heart after myocardial injury.
Chronic heart failure (HF) occurs when the heart is unable to pump enough blood to
supply the blood flow needed to maintain the circulation of the body. It is an increasing
health problem that is associated with high morbidity and mortality together with poor
prognosis. (Finegold et al. 2013 and Sanz et al. 2013)
Finding an effective animal model of atherosclerosis and myocardial ischaemia as well
as MI involving reduced left ventricle (LV) function and remodelling with long survival
is difficult to achieve. Several porcine models of CAD and MI exist and are used in
research (Hughes et al. 2003 and Lukács et al. 2012). Still, better models for translational
research are desired.
Positron emission tomography (PET) imaging of myocardial perfusion has shown high
performance for diagnosis of CAD (Schelbert et al. 1982 and reviewed in Danad et al.
2014). For example oxygen-15 labelled water ([15O]water) can be used in quantification
of myocardial perfusion (Bergmann et al. 1984) and assessment of CAD (Reviewed in
Saraste et al. 2012). Myocardial perfusion and oxidative metabolism can be measured
simultaneously in a single PET study by using a carbon-11 labelled acetate ([11C]
acetate) tracer. Further, calculation together with cardiac function measurements gives
an estimation of cardiac efficiency (Wolpers et al. 1994 and reviewed in Knaapen et al.
2007). A fluorine-18 labelled glucose analogue 2-[18F]-Fluoro-2-deoxy-D-glucose ([18F]
FDG) can be used to determine myocardial viability (Tillisch et al. 1986 and reviewed
in Knuuti et al. 2002). Viable myocardium is defined as myocardium, which does not
contract normally due to ischaemia, but has the potential to recover its function.
New radiopharmaceuticals for cardiac PET perfusion imaging are demanded.
Nitrogen-13 labelled ammonia ([13N]ammonia), rubidium-82 chloride ([82Rb]Cl) and
[15O]water are the currently used PET tracers for myocardial blood flow quantification.
These tracers have short physical half-lives and the production requires an expensive
on-site cyclotron. A longer half-life enables centred production of the tracer and delivery
for longer distances. Gallium-68 (68Ga) labelled radiopharmaceuticals have certain
advantages in preclinical research including production with relatively inexpensive
12
Introduction
germanium (68Ge)/68Ga generator and easy labelling of different molecules (Maecke et
al. 2007). Recently, promising 68Ga-labelled tracers for myocardial perfusion imaging
(MPI) have been introduced (Hsiao et al. 2009).
Because of a high mortality and poor prognosis, new treatments for HF are needed.
Positive inotropic therapy improves the cardiac performance including contractility
leading to increased pump function. A calcium sensitiser agent levosimendan has also
shown to restrict the size of MI via activation of adenosine triphosphate-sensitive
potassium (KATP) channels (Kersten et al. 2000).
In these studies we aimed to develop large animal models of CAD, myocardial ischaemia
and MI. Multimodality imaging methods were used in validation of these experimental
models together with histopathological evaluation. Also, new radiopharmaceuticals
for MPI with PET were tested. Additionally, the effect of levosimendan therapy on
myocardial infarct size and LV function were tested in a model of chronic HF.
2.
Review of the Literature13
REVIEW OF THE LITERATURE
2.1 Coronary artery disease
Atherosclerosis is a progressive inflammatory disease. Signs of atherosclerosis have
been reported already in children and even in fetuses (Stary 2000 and Napoli et
al. 1997). Several risk factors like hypertension, high cholesterol levels, diabetes,
obesity and smoking are all related to increased risk of atherosclerosis (Dahlöf 2010).
Coronary artery disease (CAD) is caused by atherosclerotic plaque formation in the
coronary artery vessel wall. Formation of an atherosclerotic plaque is a complex
process including inflammation, accumulation of lipids, cell death and fibrosis
(Hansson et al. 2006). Narrowing of coronary arteries caused by atherosclerosis can
finally lead to reduced blood flow and myocardial ischaemia. Myocardial infarction
(MI) is usually caused by advanced CAD and atherosclerotic plaque rupture.
Atherosclerosis usually develops slowly over years and is often diagnosed after the
onset of symptoms or an acute cardiac event. Ischaemic heart disease, originating
from coronary atherosclerosis, is the leading cause of death worldwide (Finegold et
al. 2013).
2.1.1 Atherosclerotic inflammation and plaque formation
Atherosclerotic plaques develop in the arterial wall in response to local endothelial
cell dysfunction and local inflammation (Libby 2012). Cardiovascular risk factors like
hypertension, hypercholesterolemia, diabetes and smoking can cause endothelial cell
dysfunction, which leads to the upregulation of adhesion molecules and inflammatory cell
recruitment (Libby et al. 2005). Atherosclerosis is initiated by the accumulation of lowdensity lipoprotein (LDL) particles into the vessel wall (Skålén et al. 2002). Oxidation of
LDL leads to the expression of leukocyte adhesion molecules like vascular cell adhesion
molecule-1 (VCAM-1), which lead to the infiltration of monocytes and T lymphocytes
and, in combination with leukocytes, causes the secretion of chemokines such as
tumour-necrosis factor alpha (TNFα), interleukins (ILs), monocyte chemoattractant
protein-1 (MCP-1) and matrix metalloproteinases (MMPs) (Libby 2002). Monocyte
differentiation into macrophages is induced by macrophage-colony stimulating factor
(M-CSF). Subsequently, scavenger receptors (SRs) and Toll-like receptors (TLRs) are
upregulated. Formation of foam cells is mediated by SR, whereas TLR initiated signal
cascades lead to the activation of inflammation (Figure 1) (Yan et al. 2007). Inflammatory
processes and accumulation of lipid droplets lead to initial atherosclerotic changes called
fatty streaks and later formation of advanced plaques and atheromas (Hansson et al.
2006).
14
Review of the Literature
Blood monocyte
Adhesion molecule
VCAM‐1
Arterial lumen
Arterial intima
SR
T lymphocyte
TLR
Oxidised LDL
Monocyte becoming
intimal macrophage
Macrophage
Foam cell
Chemokines
(e.g., TNFα, ILs, MCP‐1, MMPs)
Figure 1. Inflammatory processes related to atherosclerosis.
2.1.2 Coronary artery stenosis and plaque rupture
The earliest stage of progressive atherosclerosis is intimal thickening, which is defined
as an increased number of intimal smooth muscle cells. This continuous inflammatory
process leads to the formation of a fibroatheroma, a lesion with a necrotic core and an
overlying fibrous cap. Progressive atherosclerosis leads to more advanced plaques and
narrowing of vessel lumen. Over time, atherosclerosis can cause myocardial ischaemia
by the narrowing of the vessel lumen or plaque rupture. Rupture often develops a
luminal thrombus, which is a common cause of acute MI and sudden death (Virmani
et al. 2000).
2.2 Ischaemic cardiomyopathy
2.2.1 Myocardial ischaemia
Myocardial ischaemia occurs when the supply of oxygen no longer meets the requirement
of myocardial demand. Myocardial ischaemia is caused by the limited amount of blood
supply into myocardial tissue. The blood supply of heart is organised through coronary
circulation and CAD is the most common cause of chronic myocardial ischaemia or
infarction (Sanz et al. 2013). Insufficient blood flow can lead to regional myocardial
dysfunction and later on to cardiac remodelling and HF.
Under normal aerobic conditions, fatty acids supply 60% to 90% of the energy for
adenosine triphosphate (ATP) synthesis. Approximately 10% to 40% of the energy
comes from oxidation of pyruvate and only small amount (3%) is derived by glycolysis
(Stanley 2004). Partial reduction in coronary blood flow shifts cardiomyocytes to utilise
Review of the Literature15
more glucose. Glucose metabolism needs 10% less oxygen than the oxidation of fatty
acids. Still, the most of the energy (50-70%) is derived from the oxidation of fatty acids
(Figure 2) (Stanley 2001). Critical myocardial ischaemia results in increase of anaerobic
glycolysis leading to production of lactate. The myocyte swells due to imbalance of
intracellular homeostasis, which can lead to ischaemic cell death and a decrease in
contractile work.
Aerobic conditions
Ischaemic conditions
Glycolysis
Fatty acids
3%
60-90%
Lactate
Pyruvate
Glycolysis
ATP
ADP
Pyruvate
Lactate
10-40%
Contraction
ATP
Fatty acids
50-70%
30-40%
O2
Contraction
ATP
Mitochondrion
ADP
Mitochondrion
ADP
Figure 2. Myocyte energy metabolism under normal aerobic and ischaemic conditions.
2.2.2 Ischaemic preconditioning
Ischaemic preconditioning refers to the myocardial protection occurring after brief
periods of sublethal ischaemia. It has been linked to reduced MI size and increased
survival. Protective effects start immediately after coronary occlusion following
reperfusion (first window) (Murry et al. 1986). The first window of protection is
activated through opioid, adenosine, bradykinin or alpha1-adrenergic receptors
lasting 4 to 6 hours (Figure 3) (Rodrigo et al. 2008). Activation of protein kinase
C (PKC) leads to activation of mitochondrial KATP channel. The second window of
protection has been demonstrated 24 hours after the preconditioning lasting up to
72 hours (Buja 2005 and Das et al. 2008). The second window is activated through
nuclear factor kappa-B (NF-κB) (Figure 3). Preconditioning reduces the energy
demand of the myocardium leading to preserved myocardial function and reduction
of arrhythmias (Hagar et al. 1991). The exact mechanisms of this preconditioning
effect remain unclear.
16
Review of the Literature
Ischaemia/reperfusion
Receptor activation
(opioid, adenosine, bradykinin)
PI3K
Cardiac nerves
(α1‐adrenergic receptors)
2+
PKC
Endotoxins
PKC, MAPK
TK, JAK/STAT
Ca
Akt
Exercise
NO
NF‐κB
GSK‐3β
Mitochondrial KATP channel
Transition pore
Oxygen radicals
First window
iNOS, COX‐2, MnSOD
HO‐1, aldose reductase
Second window
Figure 3. Mechanisms of the first and second window of ischaemic preconditioning.
2.2.3 Myocardial infarction
Myocardial infarction (MI) is caused by severe and prolonged ischaemia. The existence
of coronary collateral flow is one determinant of infarct size. Necrosis occurs first in the
subendocardial region. The necrotic area expands progressively transmurally, i.e., affecting
the entire thickness of the wall within time (Reimer et al. 1979). The major cause of acute
MI is coronary atherosclerosis with luminal thrombus. MI without atherosclerosis is rare.
A prolonged period of insufficient blood flow leads to MI. Wound-healing involves fibrosis
formation and MI scarring. Inflammatory cells, mainly macrophages, initially infiltrate
into the necrotic myocardium. Interstitial fibroblasts are transformed into myofibroblasts
by transforming growth factor-β1 (TGF-β1). Myofibroblasts express alpha-type smooth
muscle actin and contain angiotensin-converting enzyme and matrix metalloproteinases.
TGF-β1 and angiotensin II are responsible for accumulation of collagen. Changes in the
myocardial extracellular matrix are very rapid and formation of collagen in the infarcted
area is visible two to three days after MI. Infarcted myocardium thins and is replaced with
non-functioning collagen-rich scar tissue (Swynghedauw 1999 and Bujak et al. 2007).
2.2.4 Cardiac remodelling
Myocardial remodelling includes all those changes that occur after MI to compensate for
the loss of a functional myocardium. Remodelling is initiated by increased mechanical
Review of the Literature17
stretch due to MI scarring. A major determinant for infarct expansion and remodelling
is the transmural extent of necrosis. Complex neurohumoral remodelling processes
usually lead to viable cardiac tissue hypertrophy, fibrosis and cell death. After MI,
systolic function will be impaired due to the loss of contractile myocardium. This leads
to increased end-systolic volume, increased heart size and increased diastolic filling
pressure (Swynghedauw 1999). Elevated myocardial wall stress will be normalised by
hypertrophy according to Laplace’s law. The wound healing and remodelling processes
after MI involves both the infarcted and non-infarcted myocardium. Fibrosis is more
related to scar formation when it is replacing necrotic tissue but also to remodelling
occurring interstitially in non-infarcted remote tissue. Fibrosis in the remote myocardium
can be induced by vasoactive peptides and hormones like angiotensin II and by
mechanical pressure overload (Heusch et al. 2014).
2.2.5 Heart failure
HF occurs when the heart is unable to pump enough blood to maintain normal
circulation. It is a consequence of an abnormality in cardiac structure, function, rhythm
or conduction. The most common causes of heart failure are myocardial infarction
and hypertension (McMurray et al. 2005). HF can appear acutely especially in the
case of MI. Most clinical signs are non-specific. Electrocardiography (ECG) can be
used to measure pathological changes in the electrical conduction system of the heart.
Echocardiography is widely used to demonstrate cardiac dysfunction. Other imaging
modalities, such as computed tomography (CT), PET and magnetic resonance imaging
(MRI) are used increasingly in the evaluation of HF. Despite recent advances in
pharmacological and device therapies, prognosis of HF is often poor: 30 to 40 percent
of patients die within a year and 60 to 70% die within 5 years of diagnosis (McMurray
et al. 2005).
Energy metabolism is disturbed in HF. Contractile performance is decreased together
with reduction of ATP, phosphocreatine and creatine kinase concentration (Liao et al.
1996). Oxygen deprivation results in impaired relaxation and contraction. Stunning can
be present after prolonged ischaemia as decreased contraction persisting for hours after
the return of blood flow. During hibernation, blood flow is chronically limited and the
myocardium contraction fails. Inadequate blood flow leads to MI, LV remodelling and
further HF (Vanoverschelde et al. 1993).
The oxygen requirement of the heart can be increased in HF due to increased cardiac
work, myocardial mass, increased myocardial wall stress and wasted contractile energy.
After MI, neurohumoral activation and myocardial load and stretch are increased. It
leads to myocardial growth response inducing myocardial hypertrophy and myocardial
remodelling. The final outcome is an increased cardiac energy expenditure and
myocardial cell death through apoptosis and necrosis leading to HF (Figure 4) (Heusch
et al. 2014).
18
Review of the Literature
Myocardial infarction
Load, stretch
Cardiac output
Load, stretch
Neurohumoral activation
Myocardial
growth
response
Cardiac
energy
expenditure
Myocardial
hypertrophy
Cell
elongation
Remodelling
Apoptosis
Myocyte necrosis
Myocardial cell death
Heart failure
Figure 4. Mechanisms leading to myocardial remodelling and HF.
2.3 Experimental models of coronary artery disease
Atherosclerosis research using human samples is difficult because of the slow and
unnoticeable development of the disease. Tissue samples are also difficult or impossible
to obtain from humans. Therefore, animal models are needed. Several small animal
atherosclerosis models have been developed by genetic engineering, e.g., modifying
genes coding the LDL receptor or apolipoproteins (Heinonen et al. 2007). Despite several
benefits, small animals have some restrictions like small size and differences in anatomy
and physiology compared to humans. Large animal models are better suitable for studying
coronary arteries, which are difficult or even impossible to study by using small animals.
Also, achieving a more human-like size, anatomy and physiology is critical in translational
research where basic research advances can be easily transferred onto a clinical stage.
Rabbit models have been widely used in cardiovascular research. Watanabe rabbits have
a high serum level of LDL-cholesterol and are prone to produce atherosclerosis within
a reasonable time when fed with an atherogenic diet (Aliev et al. 1998). A pig model
Review of the Literature19
is considered very appropriate because the anatomy and physiology of the heart and
coronary vasculature are very similar to that of humans (Bertho et al. 1964, Kamimura
et al. 1996, Kassab et al. 1997 and Swindle et al. 2012). The anatomy of the pig heart
is nearly similar than that of the human heart. The LV wall is thicker than in human
heart. Only two pulmonary veins drain into the left atrium. The left azygous vein drains
into the coronary sinus, but otherwise the coronary circulation is identical with humans
(Weaver et al. 1986, Crick et al. 1998 and Swindle et al. 2012).
Pig models are used for characterising the role of monocytes during early atherosclerosis
as well as smooth muscle and endothelial cell proliferation in plaque development
(Gerrity 1981a, Gerrity 1981b and Gerrity et al. 1985). Atherosclerotic lesions are
mainly located similarly as in humans in the proximal part of coronary arteries (Gerrity
et al. 2001 and Thim et al. 2010). The morphology and progression of plaques in pig are
similar to that seen in humans. Also, blood glucose, cholesterol and lipoprotein levels
and metabolism are similar to those of humans.
2.3.1 Large animal models of atherosclerosis
A large animal model mimicking human-like coronary atherosclerosis is lacking,
but highly needed for translational research approaches. Farm pigs with familial
hypercholesterolemia develop atherosclerosis in two to three years (Thim et al. 2010).
In that case, animals are usually weighing more than 200 kg and the extensive housing
costs of pigs are expensive. Atherosclerosis can be induced by different techniques. The
most relevant techniques with characteristics are presented in Table 1.
Normally farm pigs are naturally very resistant to develop inherent atherosclerosis.
Early atherosclerotic lesions, mainly fatty streaks, are possible to achieve by
hypercholesterolemia that is induced by an atherogenic diet containing high amounts
of saturated fatty acids and cholesterol. Blood total cholesterol level increases from a
normal level of 2 mmol/L to 5 mmol/L with a diet containing 2 percent cholesterol and
17 percent to 20 percent fat (Artinger et al. 2009 and Pueyo Palazón et al. 1998). With
additional sodium cholate in the diet (1.5%), it is possible to achieve more advanced
atherosclerotic plaques including fibroatheroma and cholesterol crystals with 24 to 41
percent coronary artery stenosis (Thorpe et al. 1996).
The formation of atherosclerosis can be accelerated by inducing diabetes by using
streptozotocin to destroy insulin-secreting beta cells in pancreatic tissue and feeding
pigs an atherogenic diet containing a high amount of cholesterol (1.5%) and lard (15%)
(Gerrity et al. 2001). In that model both plasma glucose and cholesterol levels are
possible to maintain near 20 mmol/L leading to more advanced atherosclerotic plaque
formation including a well-developed fibrous cap overlying necrotic lipid core and
coronary artery stenosis as high as 98 percent (Gerrity et al. 2001). Plaque development
can also be controlled in these models using statin or angiotensin converting enzyme
inhibitor therapy (Chatzizisis et al. 2009).
20
Review of the Literature
Genetic models have also been demonstrated. In a minipig model that has a LDL
receptor mutation, atherosclerosis was successfully induced by an atherogenic diet and
mechanical injury of vessel wall (Thim et al. 2010).
Pigs having mutant alleles for apolipoprotein B develop spontaneous
hypercholesterolemia. Early atherosclerotic changes are visible during the first year. At
two years of age, advanced plaques with a necrotic core with fibrous cap and stenotic
lesions exist. By three years of age, complicated lesions and rupture are common.
(Prescott et al. 1991)
Rapacz familiar hypercholesterolemia pigs develop atherosclerotic lesions mainly in
peripheral arteries and are thus more relevant in studies related to peripheral arterial
disease (Bahls et al. 2011).
D374Y gain-of-function mutations in the proprotein convertase subtilisin/kexin type 9
(PCSK9) gene lead to autosomal dominant hypercholesterolemia and atherosclerosis
in humans. Pigs with DNA transposition of a human PCSK9 gain-of-function mutant
represent an atherosclerosis model of familial hypercholesterolemia. (Al-Mashhadi et
al. 2013)
Ossabaw swine represents a metabolic disease model representing increased blood
glucose and impaired insulin sensitivity. Early atherosclerotic changes develop over
time. (Dyson et al. 2006)
To accelerate CAD development, an oversized angioplasty balloon can be used to
induce mechanical vessel wall injury. Histological changes are usually occur at
the very early stage, but also advanced atherosclerotic lesions including neointima
expansive remodelling and fibroatheroma have been observed in these models (Thim
et al. 2010).
Coronary occlusion can be induced with a mechanical occluder to simulate plaque rupture,
a complication of atherosclerosis. An ameroid constrictor has been used in atherogenic
diet fed pigs in studies related to mechanisms after the rupture, e.g., collateral formation
(Matyal et al. 2012).
The limitations of pig models of CAD include the lack of spontaneous development of
atherosclerosis and that formed plaques remain at the initiation state without stenosis
and rupture leading to MI. Also, atherosclerosis can only be induced over prolonged
periods of time, which leads to increased costs and difficulties when handling heavy
animals.
Review of the Literature21
Table 1. Characteristics of pig models of atherosclerosis.
Technique
Characteristics
Hypercholesterolemia
Early atherosclerotic lesions, fatty streaks,
intimal thickening, low-grade stenosis
Hypercholesterolemia + hyperglycemia
Genetic modifications
Metabolic disease
Vessel wall injury
Reference
Artinger et al. 2009, Turk et al. 2005,
Pueyo Palazón et al. 1998, Thorpe
et al. 1996, Kim et al. 1993 and
Shimokawa et al. 1988
Gerrity et al. 2001, Wilensky et al.
2008, LiFeng Zhang et al. 2003,
Advanced atherosclerotic lesions, necrotic
Hamamdzic et al. 2010, Mohler et al.
lipid core, well-developed fibrous cap, high2008, Artinger et al. 2009, McDonald
grade stenosis
et al. 2007, Dixon et al. 1999 and
Chatzizisis et al. 2009
Prescott et al. 1991, Hasler-Rapacz et
al. 1998, Hasler-Rapacz et al. 1995,
Spontaneous hypercholesterolemia, fatty
Prescott et al. 1995, Tellez et al. 2010,
streaks, advanced plaques containing necrotic
Bahls et al. 2011, Granada et al. 2011,
core, calcification, neovascularization,
Thim et al. 2010, Al-Mashhadi et al.
hemorrhage and rupture.
2013 and Thim, Hagensen, WallaceBradley, et al. 2010
Increased blood glucose, insulin resistance,
Dyson et al. 2006, Neeb et al. 2010
early atherosclerotic changes
and Kreutz et al. 2011
Tellez et al. 2011, Worthley et al. 2000,
Early atherosclerotic-like lesions, eccentric
Worthley et al. 2000, Mihaylov et al.
fibrocellular plaques, increased intima-media
2000, Carter et al. 1996, Shimokawa et
ratio
al. 1985 and Thim et al. 2010
Matyal et al. 2012, Matyal et al.
Regional ischaemia/infarction
2013, Lassaletta et al. 2012 and
Robich et al. 2011
Hypercholesterolemia + mechanical
coronary occlusion
Intramural injection of cholesteryl Lipid-containing inflammatory lesions
linoleate
Granada et al. 2005
2.4 Experimental models of ischaemic cardiomyopathy
The most used experimental models for ischaemic cardiomyopathy are mouse and rat.
Rodents have many advantages like the availability, cost-effectiveness, easy handling
and transgenic strains. However, there are significant differences in the anatomy and
physiology of the heart and circulatory system between rodents and humans.
Dog models of myocardial ischaemia have been successfully used to study mechanisms
behind MI and later myocardial remodelling and HF (Reimer et al. 1977, Reimer et al. 1979,
Przyklenk et al. 1986 and Jugdutt et al. 1979). The pre-existing collateral circulation makes it
difficult to induce large myocardial ischaemia or infarction in dogs (Jugdutt et al. 1979). The
absence of an existing collateral network enables studies related to myocardial adaptation to
ischaemia in pigs. The pig heart and circulatory system is very close to that of humans and is
therefore suitable as a humanoid model of heart diseases (Swindle et al. 2012).
Several pig models having different cardiomyopathies have been introduced and can
be used in studies of mechanisms of heart failure and cardiac remodelling. Pressure
22
Review of the Literature
overload induced by aortic stenosis can be used to cause LV hypertrophy through adaptive
mechanisms that normalise to increased LV wall stress (Gelsomino et al. 2013). This
situation contributes to the preservation of normal LV function and excessive collagen
accumulation (Dixon et al. 2009). LV hypertrophy is a significant contributor of HF.
Reduced LV relaxation and filling together with increased LV stiffness creates pressure
overload models well-suited for the investigation of HF. Volume overload models can
be used to mimic the clinical situation of mitral valve regurgitation. A disease model can
be induced by chordal rupture of the mitral valve. Volume overload produces cellular
level abnormalities causing decreased myocyte contractile function finally leading to
HF. (Dixon et al. 2009)
Doxorubicin-induced HF is another model to study global HF. Intravenous injection
of cardiotoxic doxorubicin rapidly progresses to systolic and diastolic dysfunction and
ECG abnormalities (Torrado et al. 2011).
Dilated cardiomyopathy is defined as LV dilatation resulting in increased LV wall stress.
Pacemaker-induced chronic tachycardia has been used to induce dilated cardiomyopathy
and HF since 1962 (Dixon et al. 2009). Nowadays modern telemetry systems can be used
in these models for chronic monitoring of changes in several hemodynamic parameters
simultaneously (Choy et al. 2014).
2.4.1 Large animal models of chronic myocardial ischaemia
Myocardial ischaemia is possible to induce in several ways by causing limited blood flow
in coronary artery with stenosis. In surgical open-chest models, coronary artery stenosis
is induced by an occluder placed around the coronary artery. An ameroid constrictor
placed around the coronary artery induces gradually an increasing reduction in blood
flow leading finally to total occlusion of coronary artery. An adjustable occluder or shunt
applications can be used to obtain different levels of stenosis. Mechanically working
hydraulic, screw type or inflatable occluders can be used to achieve adjusted blood flow.
For studies concerning myocardial angiogenesis, energy metabolism and other cellular
mechanisms under chronic myocardial ischaemia, a constantly limited blood flow is
desired. Fixed-diameter occluders produce constant stenosis and permanently limited
blood flow. Delran stenosis, C-shape occluder or ligations are mostly used to produce
fixed stenosis.
Myocardial ischaemia is possible to induce percutaneously by using invasive angiographic
methods. In closed-chest models, ischaemia can be induced with an angioplasty balloon,
intracoronary flow-reducer, stent or by vessel wall injury. An angioplasty balloon can
be used to create acute myocardial ischaemia whereas chronic ischaemia is possible to
induce by stents or vessel wall injury.
Chronic myocardial ischaemia leads to deteriorated LV function due to decreased
contractility. Regional ischaemia is often seen as decreased wall motion, perfusion and
Review of the Literature23
oxygen consumption. Apoptosis, glucose utilisation and lactate accumulation are usually
increased. Different techniques with characteristics and selected references are listed in
Table 2.
Changes in vessel geometry and intracoronary artificial occluders predispose to
thrombosis leading to acute MI. Anticoagulant therapy has been successfully used to
prevent premature coronary obstruction.
Table 2. Characteristics of pig models of chronic myocardial ischaemia.
Technique
Ameroid
constrictor
Carotid coronary
shunt
Characteristics
Reference
Reduced left ventricular function, reduced
Caillaud et al. 2010 and Giordano et
blood flow
al. 2013
Increased K+ concentration, decreased pH, ST
Watanabe et al. 1987
changes
Fallavollita et al. 1999, Fallavollita
et al. 1997, Fallavollita, Lim, et al.
2001, Fallavollita, Logue, et al. 2001,
Regional LV motion abnormality, reduced
Fallavollita 2000, Fallavollita 2002,
Fixed-diameter
resting perfusion, hibernating myocardium,
Fallavollita et al. 2002, Fallavollita et
occluder
reduced ejection fraction (EF), increased
al. 2005, Lim et al. 1999, Hardt et al.
apoptosis, reduced flow reserve
2001, McFalls et al. 2006, McFalls et
al. 1997, Mills et al. 1994, Bloor et al.
1984, Ishikawa et al. 2011 and Heilmann et al. 2006
Regional LV motion abnormality, decreased Thomas et al. 1999, Kim et al. 2001,
wall-thickening, reduced perfusion, impaired St. Louis et al. 2000, Hughes et al.
Ca2+ handling, depletion of contractile materi- 2001, Chen et al. 1996, Chen et al.
Adjustable
al, glycogen accumulation, increased amount 1997, Lai et al. 2000, Liedtke et al.
occluder
of mitochondria, decreased oxygen consump- 1995, Chen et al. 1997, Liedtke et al.
tion, increased glucose consumption, lactate 1994, Bolukoglu et al. 1992, Cason et
production, reduced left ventricular EF, inal. 1991, Galiuto et al. 2002 and Sascreased apoptosis, decreased flow reserve
sen et al. 1988
Angioplasty
Bamberg et al. 2012 and Mahnken et
Regional perfusion defect
balloon
al. 2010
Gewirtz et al. 1981, de Groot et al.
2011, von Degenfeld et al. 2003,
Intracoronary flow- Regional perfusion defect, impaired myocar- Johnson et al. 1998, Gewirtz, Brautireducer
dial function, increased glucose utilisation
gan, et al. 1983, Gewirtz, Williams, et
al. 1983, Kraitchman et al. 2000 and
Kraitchman et al. 2002
Wu et al. 2011, Wu et al. 2010, Bito et
Decreased ejection fraction, hibernation, deal. 2004, Szilárd et al. 2000 and HorCopper stent
creased wall-thickening
stick et al. 2009
In-stent neointimal
Hemetsberger et al. 2008 and Johnson
Over 40% stenosis after 1-month follow-up
hyperplasia
et al. 2000
Grinstead et al. 1994, Schwartz et al.
Coronary artery
Neointimal thickening, over 50% stenosis
1990, Schwartz et al. 1992 and Anderinjury
after 1-month follow-up
sen et al. 1996
24
Review of the Literature
2.4.2 Large animal models of acute ischaemia-reperfusion injury and infarction
Myocardial injury leading to impaired LV function and HF can be induced by
prolonged episodes of myocardial ischaemia followed by reperfusion. Temporal
occlusion by angioplasty balloon is the most commonly used method to occlude
coronary artery completely but only for a short period of time. Regional ischaemia
occurs immediately after inflating the balloon. Deflating the balloon leads to
reperfusion. Severity of ischaemia-reperfusion injury depends on the duration of
ischaemia following reperfusion. Different techniques and characteristics are
presented in Table 3.
A cardiopulmonary bypass model can be used to mimic the situation in open heart
surgery or transplantation. An animal is first connected to a heart-lung machine.
The heart is arrested with cardioplegia and the remaining body is perfused by using
cardiopulmonary bypass. Global myocardial ischaemia-reperfusion injury can be
studied after weaning from a heart-lung machine. Despite that the heart is wellprotected with cardioplegia, damage of muscular fibres, mitochondrial swelling and
intracellular oedema can be observed with transmission electron microscopy (Hong
et al. 2013). Hemodilution, volume loading as well as cytokine and catecholamine
surge are often induced after cardiopulmonary bypass and reperfusion (Olson et al.
2012).
The clinical situation with coronary artery occlusion following the resolution of clots
and reperfusion can be modelled by regional ischaemia-reperfusion injury induced
with temporal occlusion of the coronary artery surgically with clamping and ligation
or percutaneously with angioplasty balloon. An ischaemic period of 30 to 120 minutes
seems to be well-tolerated. Ventricular extrasystoles, non-sustained tachycardia and
ventricular fibrillation can occur during ischaemia. Myocardial stunning is related to
inadequate contractility of cardiomyocytes after an ischaemic condition following
reperfusion and is possible to study with these models. MI is usually observed in these
models without protective cardiac arrest.
Cardiac arrest has been caused also by using alternative methods like asphyxia
induced by endotracheal tube clamping, electric fibrillation or placement of
intracoronary ball.
Review of the Literature25
Table 3. Characteristics of pig models of acute ischaemia-reperfusion injury and infarction.
Technique
Characteristics
Cardiopulmonary
bypass
Decreased LV contractility, function and
blood pressure, increased myocyte apoptosis,
coronary blood flow and troponin levels, myofibril damage and neutrophil infiltration
Coronary clamping
Stunning, decreased LV function and blood
pressure, increased heart rate, MI
Coronary ligation
Decreased LV function, increased apoptosis
and oxidative stress, MI
Balloon occlusion
Decreased LV contractility, function, increased apoptosis, MI
Asphyxia
Decreased LV contractility and function
Electric fibrillation Elevated cardiac enzymes
Intracoronary ball Elevated cardiac enzymes, MI
Reference
Hong et al. 2013, Olson et al. 2012,
Shinohara et al. 2011, Salminen et
al. 2011, Abdel-Rahman et al. 2009,
Banz et al. 2008, Jormalainen et al.
2007, Malmberg et al. 2006, Vähäsilta
et al. 2005 and Lim et al. 2005
Díez et al. 2013, Aarsæther et al. 2012
and Sala-Mercado et al. 2010
Xiang-dong Li et al. 2013, Doganci et
al. 2012, Meyer et al. 2013, Skyschally
et al. 2013, Chinda et al. 2013, Kanlop
et al. 2011, Arslan et al. 2012, Gelsomino et al. 2012, Gelsomino et al. 2011,
Oyamada et al. 2010, Sodha et al.
2009, Osipov et al. 2009, Metzsch et
al. 2006 and Garcia-Dorado et al. 1987
Hashizume et al. 2013, Duran et al.
2012, Ogura et al. 2012, Wheeler et al.
2012, Barallobre-Barreiro et al. 2012,
Lu et al. 2013, Wojakowski et al. 2012,
Chatziathanasiou et al. 2012, Bhindi et
al. 2012, Poulsen et al. 2011, Dash et al.
2011, van der Pals et al. 2010, Wiggers
et al. 1997, Silva et al. 2009, Boekstegers et al. 2002 and Hinkel et al. 2013
Lin et al. 2013
Bertsch et al. 2001 and Bertsch et al.
2000
Näslund, Häggmark, Johansson,
Marklund, et al. 1992 and Näslund,
Häggmark, Johansson, Pennert, et al.
1992
2.4.3 Large animal models of chronic myocardial infarction, remodelling and heart
failure
A large animal model of chronic MI and HF is crucial in translational research to study
complex mechanisms underlying ischaemic heart diseases. Based on literature searches,
the most common way to induce MI is to use an angioplasty balloon to occlude coronary
artery totally for a temporal period of time following reperfusion. Coronary artery
ligation, ameroid constrictor and embolisation are also widely used methods. Different
techniques and characteristics are presented in Table 4.
MI can be induced percutaneuosly by angioplasty balloon. The coronary artery is
catheterised under X-ray angiography. An angioplasty balloon is placed into the
coronary artery and inflated to occlude the vessel totally. After the desired occlusion
time, the balloon is deflated and removed. The severity of MI depends on the occlusion
26
Review of the Literature
time and the balloon location. A 60-minute occlusion time produces a large transmural
MI. Occlusion of 30 minutes does not produce myocardial necrosis and a 45-minute
occlusion produces only a small necrotic area (Garcia-Dorado et al. 1987). Ischaemic
preconditioning can be induced with repetitive balloon occlusions (Yang et al. 2011).
Coronary artery ligation and ameroid constrictor placement requires surgery. Usually
thoracotomy is enough for accessing coronary arteries. A direct view of the left anterior
descending coronary artery (LAD) is possible to achieve by dissecting skin and muscle
between the third and fourth ribs. A spreader needs to be used to separate the ribs. After
dissecting pericardium, LAD is visible.
Permanent ligation of the coronary artery leads to acute MI following LV remodelling
and worsening of LV function (Huang et al. 2010 and J. Zhang et al. 1996). Infarction
can also be induced by temporal coronary artery ligation following reperfusion (So et
al. 2012).
An ameroid constrictor placed around a coronary artery induces gradual occlusion
leading usually to MI. Complete coronary occlusion is necessary in research related to
MI and for example, collateral growth.
Induction of MI by inducing embolisation with thrombogenic material like embolisation
coils, intracoronary ethanol or a gelatine sponge leads to very similar MI and later
changes than coronary ligation (Gálvez-Montón et al. 2014).
The size of MI should be large enough to induce remodelling processes. Distal and
midpoint ligation of LAD leads to a MI size of 10 percent and 14.9 percent, respectively
(Munz et al. 2011). MI sizes of 12.8 percent and 23.8 percent are reported to be achieved
by ligating one-third of the LAD from the apex and below the second diagonal branch,
respectively (Huang et al. 2010). Over 25 percent MI of LV is possible to achieve with
proximal occlusion of LAD whereas proximal left circumflex artery (LCX) occlusion
induces MI covering 20 percent of LV (Teramoto et al. 2011, Munz et al. 2011 and Roth
et al. 1987). High mortality rates have been reported if the MI size exceeds 25% of the
LV (Kamimura et al. 1996). Sudden cardiac deaths are reported to be in relation to fatal
arrhythmias due to intolerance of ischaemia (Fallavollita et al. 2005). The ligation of a
distal part of coronary artery demonstrates reasonable survival rates but only small MI.
Gradual coronary artery occlusion induced using an ameroid constrictor has interestingly
led to very small MI and an aggressive collateral development may be the cause (Roth
et al. 1987). Combining the distal coronary ligation and proximal ameroid constrictor
demonstrates large MI (>25% of LV) with clear signs of remodelling. End-diastolic and
end-systolic volumes were markedly larger and EF lower in MI group when compared
to controls. Hypertrophy and fibrotic changes can visualised by histology in MI group.
Also, a survival rate at the 4-month time point was reported to be as high as 75 percent
(Teramoto et al. 2011). A study configuration with only a proximal ameroid constrictor
produced a survival rate of 30 percent. The pig heart is very sensitive to acute ischaemia
Review of the Literature27
and sudden cardiac deaths can occur (Fallavollita et al. 2005). The mechanism of
inhibition of sudden cardiac death with distal coronary ligation is not well-known. One
hypothesis is that distal ligation provides a preconditioning effect through ischaemia
and small MI as demonstrated by several studies (Murry et al. 1986, Kuzuya et al.
1993 and Hagar et al. 1991). Also, gradual occlusion may adapt the myocardium to
tolerate ischaemia. A chronic HF model with impaired LV systolic function including LV
dilatation, reduced EF and cellular evidence of LV remodelling offers new possibilities
in translational research.
Table 4. Characteristics of pig models of chronic MI, remodelling and HF
Technique
Characteristics
Coronary ligation
Transmural MI, impaired LV function, LV
dilatation, increased apoptosis
Ameroid
constrictor
Transmural MI, impaired LV function, LV
dilatation, impaired LV wall motion and increased myocyte hypertrophy and interstitial
fibrosis in the non-infarcted remote tissue,
rapid collateral development
Angioplasty
balloon
Transmural MI, impaired LV function, LV
dilatation
Embolisation
Transmural MI, impairer LV function and
wall motion, LV dilatation
Reference
Garcia-Dorado et al. 1987, Jiang et
al. 2014, Zhu et al. 2013, Prescimone
et al. 2013, Munz et al. 2011, Kuster
et al. 2011, So et al. 2012, Qu et al.
2012, Sahul et al. 2011, Weiss et al.
2010, Huang et al. 2010, Cho et al.
2008 and Shuros et al. 2007
Giordano et al. 2013, Kawamura et
al. 2012, Shudo et al. 2011, Teramoto
et al. 2011, Barandon et al. 2010,
Schneider et al. 2010, Tuzun et al.
2010, Pätilä et al. 2009, Christian et
al. 2008, Ikonen et al. 2007 and Roth
et al. 1987
Xiaorong Li et al. 2014, Tanaka et
al. 2014, Sheriff et al. 2014, Vilahur et al. 2014, Varga-Szemes et al.
2014, Pavo et al. 2014, van Hout et
al. 2013, Koudstaal et al. 2013, Yan
Chen et al. 2013, Duran et al. 2012,
Yang et al. 2011, Pleger et al. 2011,
Angeli, Amabile, Shapiro, et al.
2010, Angeli, Amabile, Burjonroppa,
et al. 2010, Lautamäki et al. 2009,
Pérez de Prado et al. 2009, Holz et al.
2009, Brødløs et al. 2009, Krombach
et al. 2005, Garcia-Dorado et al. 1987
and Abegunewardene et al. 2009
Gálvez-Montón et al. 2014, GálvezMontón et al. 2013, Biondi-Zoccai
et al. 2013, Fish et al. 2013, Saeed et
al. 2013, Song-Yan Liao et al. 2010,
Peukert et al. 2009, Dib et al. 2006,
Cui et al. 2005, Dogné et al. 2005,
Waksman et al. 2004, Reffelmann
et al. 2004, Sakaguchi et al. 2003,
Crisóstomo et al. 2013, Weon Kim et
al. 2011, Joudinaud et al. 2005 and
Ohtsuka et al. 2003
28
Review of the Literature
2.5 Imaging of coronary artery disease and ischaemic cardiomyopathy
2.5.1 Invasive coronary artery imaging
Coronary artery stenosis, plaques and their progression can be evaluated with invasive
angiography combined with intravascular ultrasound (IVUS) and optical coherence
tomography (OCT). Calcified plaques, shear stress as well as atheroma volume and
fibrous cap thickness can be assessed with OCT (Sanz et al. 2013).
2.5.2 Molecular imaging of atherosclerotic inflammation
Several pathological processes related to atherosclerosis can be assessed with PET
imaging. Plaque formation is a response to the inflammatory process. Macrophages
are a good imaging target (Rudd et al. 2002). Also, neoangiogenesis, hypoxia and
microcalcification are associated with advanced plaque formation and are good targets
for imaging purposes (Tarkin et al. 2014).
[18F]FDG is a radiolabelled glucose analogue, which is widely used in PET imaging
of metabolic activity. [18F]FDG is taken up by the cells using glucose transporters.
Phosphorylated [18F]FDG is trapped inside the cell. Clearance from blood circulation
is rapid and trapped tracer accumulation can be visualised even with a low background
if PET imaging is performed for some time after the tracer injection. Because of high
glucose metabolism of myocardium, the analysis of the [18F]FDG uptake in coronary
arteries can be difficult due to high background activity. A low-carbohydrate, high-fat meal
prior to scanning has been introduced to suppress myocardial uptake (Wykrzykowska et
al. 2009). Increased vascular [18F]FDG accumulation indicates increased macrophage
activity in atherosclerotic inflammation (Figure 5) (Tarkin et al. 2014).
Mannose receptors (MRs) are expressed by macrophages in high-risk plaques and are
suggested to serve as more specific targets of imaging tracers (Figure 5). 18F-labelled mannose
has successfully been used in the visualisation of atherosclerotic lesions (Tahara et al. 2014).
Overexpression of somatostatin receptors in activated macrophages can be visualised
with the [68Ga]DOTATATE (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid
octreotate) tracer (Figure 5) (Rominger et al. 2010).
The translocator protein (TSPO) ligand [11C]PK11195 (N-butan-2-yl-1-(2-chlorophenyl)N-methylisoquinoline-3-carboxamide) accumulates during vascular inflammation through
increased TSPO expression in activated macrophages (Figure 5) (Laitinen et al. 2009).
Folate receptors (FRs) are expressed by activated macrophages (Figure 5). Increased
uptake of folate receptor targeting tracers in atherosclerotic plaques has been shown in
recent studies (Ayala-López et al. 2010 and Jager et al. 2014).
Scavenger receptors (SRs) are related to the macrophage differentiation into foam cells.
Radiotracers targeting scavenger receptor may help to visualise foam cells in active
atherosclerotic lesions (Bigalke et al. 2014).
Review of the Literature29
Increased expression of different adhesion molecules are related to atherosclerotic
inflammation and, for example, P-selectin and VCAM-1 are possible targets for imaging
tracers (Figure 5) (Nakamura et al. 2013 and Nahrendorf et al. 2009).
Choline takes part in cell membrane formation. Choline is believed to be accumulated
in activated macrophages and incorporated into cell membranes after phosphorylation
by choline kinase (Figure 5). Increased uptake of [11C]choline and [18F]choline in
atherosclerotic plaques has been demonstrated (Laitinen et al. 2010 and Matter et al. 2006).
Hypoxia plays an important role in atherosclerotic plaque formation (Figure 5). [18F]
fluoromisonidazole ([18F]FMISO) is used to successfully visualise hypoxia. FMISO
is a cell permeable compound, which is rapidly reoxidised and moved out from the
cell under normal oxygenation conditions. In hypoxic cells, FMISO is covalently
bound to intracellular macromolecules and remains in the cells (Mateo et al. 2014).
[18F]EF5 (2-(2-Nitro-1H-imidazol-1-yl)-N-(2,2,3,3,3-pentafluoropropyl) acetamide)
is a lipophilic compound and has better pharmacokinetic properties than FMISO.
Atherosclerotic lesions containing hypoxia have been successfully visualised with [18F]
EF5 in atherosclerotic mouse models (Silvola et al. 2011).
Formation of atherosclerotic plaques leads to local hypoxia. Hypoxia is able to induce
increased expression of integrin αvβ3, which is related to angiogenesis (Figure 5).
Elevated integrin αvβ3 expression is linked to macrophages involved in atherosclerotic
inflammation (Antonov et al. 2004). Experiments with radiolabelled tri-peptide sequence
arginine-glycine-asparagine based tracers such as [68Ga]NOTA-RGD and [18F]galactoRGD have increased tracer accumulation in atherosclerotic plaques (Paeng et al. 2013,
Laitinen et al. 2009, Beer et al. 2014 and Haukkala et al. 2009). Also, apoptosis plays
a central role in atherosclerotic inflammation and has shown to be a possible target
for molecular imaging. For instance, apoptosis-specific Annexin 5 is used to visualise
vascular inflammation (Laufer et al. 2008).
Microcalcification is related to atherosclerotic plaque development. [18F]Sodium fluoride
([18F]NaF) is an option for identifying plaques (Figure 5) (Joshi et al. 2014).
Blood monocyte
Mannose receptor
TSPO receptor
Arterial lumen
Fibrous cap
Cell membrane
Lipid core
Adhesion
molecule
Macrophage
Foam cell
Somatostatin Glucose receptor Folate uptake
receptor
Calcification
Neoangiogenesis
Hypoxia
Figure 5. Possible PET imaging targets related to atherosclerosis are macrophage infiltration,
hypoxia, neoangiogenesis and microcalcification.
30
Review of the Literature
2.5.3 Left ventricular function and structure
Echocardiography is one of the most important techniques for the detection and assessment
of myocardial ischaemia and infarction. Cardiac ultrasound is used in the detection of
the effects of myocardial ischaemia or infarction on LV function. LV size, wall thickness
and myocardial wall motion can be examined by conventional 2D imaging. Diastolic
and systolic volumes and EF of LV can be calculated using well-established methods.
Echocardiography is a valuable tool for diagnosing MI with assessment of wall motion.
MI is, in most cases, associated with regional LV motion abnormality (Greupner et al.
2012).
CT is a technique that allows for the evaluation of cardiac structure and function. When
combined with ECG gating and a contrast agent, high quality images can be obtained.
Diastolic and systolic volumes and EF of LV can be calculated using semi-automated
analysis software. LV size, wall thickness and myocardial wall motion can also be
examined easily. The advances of CT imaging include high spatial resolution (Greupner
et al. 2012).
Cardiovascular magnetic resonance imaging (CMR) is the gold standard for the
assessment of LV function (Pennell et al. 2004). The assessment of cardiac function can
be implemented using CineMRI, which is accurate in the measurement of diastolic and
systolic volumes and EF as well as cardiac mass (Scholtz et al. 2014).
2.5.4 Myocardial perfusion
Myocardial perfusion imaging (MPI) can be performed with several techniques.
Nuclear cardiology has been growing in the diagnosis and risk assessment of patients
with suspected heart disease. Single-photon emission computed tomography (SPECT)
was used in MPI since the early 1980s with thallium-201 (201Tl) and Technetium-99m
hexakis(2-methoxy-2-methylpropylisonitrile) ([99mTc]sestamibi) after approval by the
United States Food and Drug Administration (FDA) in 1990. In addition to 201Tl and
[99mTc]sestamibi, a newer widely used SPECT tracer [99mTc]tetrofosmin allows evaluation
of myocardial perfusion (Kelly et al. 1993).
PET is increasingly used in MPI. Kinetic properties of PET tracers enable more accurate
quantification of myocardial blood flow. For example, [15O]water is ideal for the blood
flow quantitation (Knuuti et al. 2009).
Cardiac MRI is becoming more commonly used in MPI with a gadolinium-based
contrast agent. First-pass perfusion can be detected. The gradient echo pulse sequence is
most commonly used to visualise perfusion. Stress perfusion MRI has a high diagnostic
accuracy in detecting coronary artery disease (Scholtz et al. 2014).
CT imaging allows also MPI with dynamic acquisition. Iodinated contrast bolus behaves
similar to gadolinium-based contrast agents used in MRI. The most recent techniques
Review of the Literature31
with 320-row CT enable MPI covering whole heart with relatively high temporal
resolution (George et al. 2012).
Myocardial perfusion can be assessed with echocardiography using gas-filled
microbubbles, nano- or microparticles as a contrast media (Seol et al. 2014).
Resting myocardial perfusion remains normal until occlusion of a coronary artery exceeds
90% (Gould et al. 1974). Stress imaging can be used to confirm perfusion defect in unclear
case. Pharmacologic stress testing is used with coronary vasodilators, e.g., adenosine
or regadenoson. Coronary blood flow will be increased three- to five-fold. Combining
rest- and stress-imaging enables calculation of myocardial flow reserve (MFR). Resting
myocardial blood flow (MBF) values can be corrected with rate-pressure product (RPP).
Changes in hemodynamics during vasodilatator infusion are possible at least with a high
dose of vasodilators used in experimental study settings. To maintain blood pressure, the α1adrenoceptor agonist phenylephrine can be used to oppose the systemic effects of adenosine,
while leaving adenosine-induced coronary vasodilation unperturbed (Sorop et al. 2008).
MPI results are usually presented using 17-segmental standardised myocardial
segmentation and nomenclature for the tomographic imaging of the heart as proposed by
the American Heart Association (Cerqueira et al. 2002).
2.5.5 Myocardial infarction and viability
[99mTc]Sestamibi is the most used SPECT tracer for assessment of myocardial viability.
Retention in myocardium is dependent on the intensity of the Na+/K+ pump in the cell
membrane and reflects the intracellular level of potassium. [99mTc]Sestamibi is taken up
by mitochondria (Allman 2013).
[18F]FDG is a widely used PET tracer for assessing myocardial viability. PET has
higher spatial resolution than SPECT. Metabolic clamping can be used for standardising
myocardial glucose utilisation. The myocardium usually uses free fatty acids (FFAs) for
energy production. Glucose and insulin administration before tracer injection switches
energy metabolism to use more glucose than FFAs. [18F]FDG is trapped inside the viable
myocyte through phosphorylation by hexokinase. The clearance from blood circulation
is fast allowing high myocardium-to-blood ratios. [18F]FDG viability imaging is often
combined with MPI using [13N]ammonia, [82Rb]Cl or [15O]water.
Previous approaches can be replaced by a dynamic imaging with carbon-11 labelled
acetate ([11C]acetate). Quantification of regional MBF and oxidative metabolism are
possible with a single study (Wolpers et al. 1997).
[15O]Water enables calculation of perfusable tissue fraction (PTF) and perfusable tissue
index (PTI). Assessment of viability has also demonstrated to be possible by using PTF
calculated by a [15O]water study (Iida et al. 2012).
32
Review of the Literature
Cardiac MRI enables viability assessment by using a gadolinium-based late enhancement
method to define the existence of the MI scar. The transmural extent of the MI scar is
possible to visualise very accurately from MRI images with high spatial resolution (Kim
et al. 2000).
Viable myocardium can be estimated by measuring the contractile reserve during stress.
Based on this, echocardiography can be used for the evaluation of viable myocardium
(Bisplinghoff et al. 2014).
2.5.6 PET imaging of myocardial metabolism
Myocardial energy expenditure can be measured with PET. FFAs are a major myocardial
energy substrate under normal conditions. Energy metabolism in failing myocytes is
altered and shifted to use more glucose. Myocardial glucose utilisation can be assessed
with [18F]FDG or [11C]glucose and fatty acid utilisation with [18F]FTHA (14(R,S)-[18F]
fluoro-6-thia-heptadecanoic acid) or [11C]palmitate (Tuunanen et al. 2011).
Oxidative metabolism is affected rapidly after MI. Myocardial oxygen consumption can
be estimated with [11C]acetate imaging. Myocardial efficiency, which is defined as the
ratio between cardiac work and myocardial oxygen consumption, is often decreased
in HF and can be measured non-invasively by combining measurement of myocardial
oxygen consumption and LV work (Bengel et al. 2000).
New specific radioligands make it possible to evaluate cellular changes related to postMI remodelling. For example the development of new blood vessels can be assessed
with RGD–based tracers targeting αvβ3 integrin in relation with neoangiogenesis (Kiugel
et al. 2014).
2.6 Positive inotropic therapy in heart failure
The calcium ion (Ca2+) has a central role in myocyte contraction. The calcium ion
makes the contraction possible by binding to troponin C, which leads to conformational
changes in troponin I. During relaxation, Ca2+ will be released from troponin C and will
be transported to sarcoplasmic reticulum or outside the cell (Figure 6). Phospholamban
regulates the storing of calcium ions into the sarcoplasmic reticulum. The Na+/Ca2+
exchanger delivers calcium ions out of the cell, which is also regulated by Na+/K+ATPase. Cyclic AMP increases the activity of protein kinase A (PKA), which leads
to phosphorylation of the Ca2+ channel and increased calcium ion influx into the cell.
Intracellular concentration of Ca2+ increases leading to contraction. Activation of
β-receptor leads to increased cyclic AMP and the activation of PKA. (Francis et al.
2014)
The PKA-dependent contractility of myocardium can be increased with β-receptor
agonists like dobutamine and dopamine or phosphodiesterase inhibitor, e.g., milrinone.
Review of the Literature33
The Na+/Ca2+ exchanger can be regulated with cardiac glycosides (digoxin). Calcium
sensitisers act by binding to troponins.
Levosimendan is a calcium sensitiser used for inotropic support in acutely decompensated
congestive HF. Levosimendan binds to cardiac troponin C and with the presence of
calcium ions stabilises the troponin conformation. Levosimendan has also a vasodilatory
effect by opening ATP sensitive potassium channels leading to vascular smooth muscle
relaxation. Unlike inotropic agents in general, levosimendan has neutral effects on
myocardial efficiency (Ukkonen et al. 2000). Inotropic and vasodilatory effects result in
an increased contractility and decreased preload and afterload. Also, the mitochondrial
ATP sensitive K+-channel-mediated cardioprotective effect is linked to levosimendan
(Papp et al. 2012).
Ca
2+
Cardiac glycosides
+
Ca
cAMP
AMP
ATP
Phosphodiesterase inhibitors
β-receptor agonists
2K
3 Na
PKA
+
Phospholamban
AC
Gs
β
Calcium sensitisers
2+
Na
Sarcoplasmic reticulum
Troponin complex
+
Troponin C
Figure 6. Mechanisms of cardiomyocyte calcium metabolism and contraction. Levosimendan
is a calcium sensitiser acting through binding to cardiac troponin C and with the presence of
calcium ions stabilises troponin conformation.
34
3.
Aims of the Study
AIMS OF THE STUDY
The purpose of this study was to develop and validate experimental large animal models
for the use in studying coronary artery disease, MI and HF using multimodality imaging
approaches. Additionally, potential radiopharmaceuticals and therapies were tested.
The specific aims of the study were:
1.
To investigate the feasibility of [18F]FDG PET imaging of inflammation in early
coronary atherosclerotic lesions in a pig model of diabetes and hypercholesterolemia
2.
To characterise cardiac remodelling in a new pig model of chronic heart failure by
assessment of left ventricular functional, metabolic and structural changes using
PET, CT and echocardiography
3.
To characterise a bottleneck stent model for chronic myocardial ischaemia and
infarction in pigs using PET imaging of myocardial perfusion
4.
To test new 68Ga-labeled tracers for PET imaging of myocardial perfusion in pigs
5.
To study the effects of long-term levosimendan therapy on myocardial infarct size
and left ventricle function after acute coronary occlusion in a pig model of postinfarction heart failure
4.
Materials and Methods35
MATERIALS AND METHODS
Finnish landrace pigs were used in the experiments. All animal experiments were
conducted in accordance with the European Union Directive for the use of experimental
animals and approved by the national Animal Experiment Board of Finland (ELLA) and
the Regional State Administrative Agency for Southern Finland (ESAVI).
4.1 Coronary artery disease model (I)
4.1.1 Experimental protocol
The formation of atherosclerotic plaques was accelerated by inducing diabetes. Diabetes
was induced by destroying beta cells using streptozotocin injections. Pigs were sedated
with ketamine (30 mg/kg intramuscularly (i.m.), Ketalar, Amgen Technology Ireland,
Dublin, Ireland) and streptozotocin 50 mg/kg (Zanosar, Pharmacia & Upjohn, Kalamazoo,
MI, USA) was injected via ear vein once a day for 3 days. In order to offset insulin release
from pancreas, glucose was given 25 g per os twice daily for 2 days. An atherogenic diet
containing either 1.5% cholesterol and 15% lard (HF, n=6) or 4% cholesterol, 20% lard
and 1.5% sodium cholate (HF+c, n=4) (Special Diet Services, Witham, UK) was started
five days after the last streptozotocin injection and was continued for 6 months. Blood
glucose and cholesterol levels were monitored regularly.
4.1.2 In vivo PET imaging of atherosclerotic inflammation
After six months of diet intervention, [18F]FDG uptake of coronary arteries was studied by
PET. Myocardial glucose uptake was suppressed with an overnight fasting and by giving
a carbohydrate free diet for two days before the PET study. PET studies were performed
by a PET and a 64 slice CT hybrid scanner (Discovery VCT, General Electric Medical
Systems, Milwaukee, WI, USA) operated in 3-dimensional mode. List-mode acquisition
for 30 minutes started 120 minutes after intravenous (i.v.) [18F]FDG injection. The acquired
list-mode data was divided into cardiac and respiratory gates and were combined as dual
gates compensating for both respiratory and cardiac motion. Results were presented as
target-to-background ratios (TBR) calculated by dividing maximum standardised uptake
value (SUV) of each coronary segment by mean SUV value of the blood.
4.1.3 Ex vivo studies
After the PET study, animals were sacrificed by i.v. injection of potassium chloride (B.
Braun Medical Oy, Helsinki, Finland). Coronary arteries were prepared and proximal
samples were collected. Samples were weighed and [18F]FDG uptake was measured with
a gamma counter (1480 Wizard 3″; PerkinElmer/Wallac, Turku, Finland). SUVs and
further vessel-to-blood ratios were calculated.
36
Materials and Methods
The coronary artery samples were mounted, frozen in isopentane mixed with dry ice and
cut into longitudinally sections. An autoradiography study with a phosphor imaging plate
(BAS-TR2025, Fuji Photo Film Co. Ltd., Tokyo, Japan) was performed. Radioactivity
distribution of the 40 μm cryosections was analysed using Fluorescent Image Analyser
(Fujifilm FLA-5100, Fuji Photo Film Co. Ltd., Tokyo, Japan).
Cryosections of 8 μm were stained with hematoxylin & eosin (HE) and Movat’s
pentachrome staining. Movat’s pentachrome stained samples were graded using a
light microscope with the following scores: 1=healthy normal vessel wall, 2=intimal
thickening, and 3=atheroma.
Tissue sections stained with HE were digitally photographed using a light microscope.
Autoradiographs and HE images were co-registered and [18F]FDG accumulation
of atherosclerotic lesions and non-atherosclerotic vessel wall as photostimulated
luminescence per square millimetre (PSL/mm2). Lesion-to-normal vessel wall ratios
were calculated for each segment.
4.2 Surgical and percutaneous myocardial infarction model (II, III)
4.2.1 Experimental protocol
Before operations or imaging studies, animals were anaesthetised by i.m. administration
of midazolam 1 mg/kg (Midazolam Hameln, Hameln Pharmaceuticals GmbH, Hameln,
Germany) and xylazine 4 mg/kg (Rompun vet, Bayer Animal Health GmbH, Leverkusen,
Germany) (II) or with atropine (0.05 mg/kg; Leiras, Helsinki, Finland) and azaperone
(Stresnil, 8 mg/kg; Janssen, Titusville, NJ, USA) (III). Animals were intubated and
ventilated mechanically. Anaesthesia was maintained with i.v. infusion of propofol
10−50 mg/kg/h (Propofol Lipuro, B. Braun Melsungen AG, Melsungen, Germany)
combined with fentanyl 4−10 µg/kg/h (Fentanyl-Hameln, Hameln Pharmaceuticals
GmbH, Hameln, Germany).
In order to induce myocardial ischaemia and infarction surgically (II), a short left anterior
thoracotomy was performed. The pericardium was opened and a complete ligation of
the distal LAD was made immediately after the second diagonal branch using a 5-0
monofilament polypropylene suture (Prolene, Ethicon, Norderstedt, Germany). After 15
minutes, the proximal LAD was prepared free and an ameroid constrictor (2.50 mm or
2.75 mm, model MRI-2.50-TI and MRI-2.75-TI; Research Instruments SW, Escondido,
CA, USA) was placed around the LAD. Arrhythmias were prevented by administration
of amiodarone (Cordarone, Sanofi-Synthelabo Ltd, Newcastle upon Tyne, UK) 8 mg/kg
perorally (p.o.) daily for 1 week before and for 2 weeks after the operation. Amiodarone 6
mg/kg i.v., metoprolol 0.2 mg/kg i.v. (Seloken, Genexi, Fontenay sous Bois, France) and
magnesium sulphate (MgSO4) 25 mg/kg i.v. (Addex-magnesium sulfate, Fresenius Kabi
AB, Uppsala, Sweden) were administered intraoperatively. Clopidogrel 3 mg/kg p.o.
(Plavix, Sanofi Winthrop Industrie S.A., Ambarès et Lagrave, France) was administered
Materials and Methods37
daily 1 day before and daily for 2 weeks after the surgery to prevent premature thrombosis
of the LAD.
A percutaneously placed intracoronary bottleneck stent was used to induce myocardial
ischaemia and infarction (III). Catheterisation was done using a GE Innova 3100IQ threedimensional (3-D) angiography device (GE Healthcare, Waukesha, WI, USA). The
bottleneck stent consisted of a bare metal stent and polytetrafluoroethylene heat shrink
tube. The other end of the tube was shaped into a bottleneck to restrict coronary blood
flow. The construct was placed into LAD with 6-F AR-2 guiding catheter. In order to
prevent arrhythmias, per oral amiodarone (Cordarone, 200 mg/day, Sanofi-Aventis, Paris,
France) and bisoprolol (Bisoprolol-ratiopharm, 2.5 mg/day, Ratiopharm, Ulm, Germany)
was started 1 week before stenting. Prior to stenting, 100 mg intravenous lidocaine (10
mg/ml, Orion Pharma, Espoo, Finland) and 2.5 ml MgSO4 (Addex-magnesium sulfate,
246 mg/ml, Fresenius Kabi, Uppsala, Sweden) was administered. One day before
stenting, per oral acetylsalicylic acid (ASA-ratiopharm, 300 mg, Lannacher Heilmittel
GmbH, Lannach, Austria) and clopidogrel (Clopidogrel Mylan, 300 mg, Mylan, Saint
Priest, France) was given. ASA (100 mg/day p.o.), clopidogrel (75 mg/day p.o.) and
enoxaparin (30 mg/day s.c.) were continued for one week to keep the stent open.
4.2.2 PET imaging of myocardial perfusion and viability
Myocardial perfusion was measured at 3 months after the ameroid constrictor placement
(II) and one week and five weeks after the bottleneck stenting (III) with [15O]water
PET. MPI was done at rest and under pharmacological stress with adenosine 200 μg/
kg/min (Adenosin Life Medical, Life Medical Sweden AB, Stocksund, Sweden). The
acquisition protocol consisted of following frames: 14 × 5 s, 3 × 10 s, 3 × 20 s, 4 × 30
s (total duration 4 min 40 s). The segmental LV myocardial blood flow was quantified
using a single-compartment model (Iida et al. 1992).
Infarcted and ischaemic myocardial regions were defined using 70% of maximum as a
threshold in [15O]water PET flow quantitation at rest and during stress, respectively (III).
Myocardial viability was assessed at five weeks after the bottleneck stenting with [18F]
FDG PET (III). Myocardial glucose utilisation was normalised with an intravenous
injection of 1 g/kg glucose and 10 IU of insulin prior tracer injection. A static 15-min
PET scan was performed 40 minutes after the injection. Viability was determined as
viable, partially viable or nonviable (relative [18F]FDG uptake 85, 67–85 and 67%,
respectively).
4.2.3 Left ventricular size and function (II)
Transthoracic echocardiography was performed for measurement of cardiac output by
pulsed-wave Doppler from the LV outflow tract (LVOT) and calculated as velocity time
integral × heart rate.
38
Materials and Methods
End-diastolic and end-systolic volumes (EDV, ESV) and EF as well as LV mass were
evaluated by contrast-enhanced CT.
4.2.4 Myocardial oxidative metabolism and efficiency (II)
Myocardial perfusion and oxidative metabolism was assessed by [11C]acetate PET
imaging. The scanning frames were as follows: 10 × 10 s, 1 × 60 s, 5 × 100 s, 5 × 120 s,
5 × 240 s (total duration 41 min). Myocardial blood flow was determined by the initial
uptake rate (k1) of [11C]acetate (van den Hoff et al. 2001). The clearance rate constant of
[11C]acetate (Kmono) was assessed reflecting myocardial oxygen consumption (Armbrecht
et al. 1990).
Myocardial infarct size was determined using a reduction of 40% as a cut-off for the k1
of [11C]acetate PET.
Global myocardial efficiency was estimated by a work-metabolic index (WMI) with the
following equation: WMI = (cardiac output) × (systolic blood pressure) / Kmono × LV mass
(Ukkonen et al. 2009). Regional efficiency of the remote non-infarcted myocardium,
Kmono, was studied in relation to the systolic wall stress using the following equation:
Efficiency = wall stress / regional Kmono.
4.2.5 Tissue samples and histology
Immediately after the imaging studies, the animals were sacrificed and heart was
prepared and sliced horizontally to four slices. After 15-min incubation in 1%
2,3,5-triphenyltetrazolium chloride (TTC) (Sigma-Aldrich, Saint Louis, MO, USA),
samples were photographed and the size of infarcted region was visually estimated.
Samples for histology were collected from the infarcted, non-infarcted remote
(inferolateral wall). Samples were mounted and frozen in isopentane mixed with dry
ice. Cryosections of 7 μm were stained with Masson’s trichrome staining. The content of
fibrosis was quantified from remote sections with the colour deconvulation method using
ImageJ software (Ruifrok et al. 2001).
4.3 New myocardial perfusion tracers (IV)
4.3.1 Study design
The suitability of four previously discovered 68Ga-labelled ligands for MPI was
tested. Results obtained with 68Ga-tracers were compared with [15O]water PET flow
quantitation. Hexadentate bis(salicylaldimine) ligands, tris(3-methoxysalicylaldimine)
(Tracer-1) and tris(3-ethoxysalicylaldimine) (Tracer-2) of bis(2,2-dimethyl-3aminopropyl)-ethylenediamine (BAPEN) and the bis(salicylaldimine) (Tracer-3) and
Materials and Methods39
bis(3-methoxysalicylaldimine) (Tracer-4) of bis(3-aminopropyl)-dimethylenediamine
(BAPDMEN) were tested in healthy pigs.
4.3.2 PET imaging and kinetic modelling of [68Ga] ligands
Myocardial blood flow (MBF) was measured first with [15O]water. Then 68Ga-tracer was
injected following 92-min scanning with following frames: 18 × 10 s, 4 × 30 s, 2 ×
120 s, 1 × 180 s, 4 × 300 s, 6 × 600 s. For analysis of MBF with 68Ga-chelates, singlecompartment model and multiple-time graphical analyses for irreversible tracer uptake
(Patlak plot) and reversible tissue uptake (Logan plot) were applied. Linear correlation
between MBF measured with [15O]water and modelling results of 68Ga-ligands was
calculated as using Pearson correlation.
4.3.3 Organ distribution
Organ samples were collected immediately after the PET imaging. Samples of whole
blood, plasma, urine, heart, lung, liver, spleen, kidney, muscle, brain, bone, bone marrow,
salivary gland and abdominal fat were prepared, weighed and measured for radioactivity
using a gamma counter (Wizard). Additionally, myocardium-to-liver, myocardium-tolung and myocardium-to-blood ratios were calculated.
4.3.4 In vitro binding to serum proteins
Binding of 68Ga-ligands to serum proteins was determined using serum obtained from
human, pig or rat. The assay was performed with the ultrafiltration method as described
earlier by Basken et al. (Basken et al. 2008) and results were expressed as an unbound
fraction (%).
4.4 Chronic levosimendan therapy for heart failure (V)
4.4.1 Study design
The pigs had a two-step occlusion of the LAD with distal ligation and proximal ameroid
constrictor. Three weeks after the surgical operation, transthoracic echocardiography was
done to visualise LV wall motion (Figure 7). Clear signs of large motion abnormality in
the LAD region was defined to be inclusion criteria. Animals with LV wall motion defect
were allocated into control group (n=18) or levosimendan group (n=7). Levosimendan
was given per orally 5 mg/kg once a day (Orion Pharma Ltd, Espoo, Finland). Intervention
was continued 8 weeks and was stopped one week before terminal imaging studies.
Imaging studies consisted of echocardiography, PET and CT. After imaging studies,
animals were sacrificed and cardiac tissue samples were collected.
40
Materials and Methods
Ligation + ameroid constrictor
Echocardiography & randomisation
Intervention ends one week
before imaging studies
3 weeks
8 weeks
Ligation + ameroid
Imaging studies
Intervention
Imaging
Blood sampling
12 weeks
Blood sampling
Figure 7. Study outline of the 8-week levosimendan intervention study where distal left anterior
descending coronary artery (LAD) was ligated and ameroid constrictor was placed in the proximal
part of LAD.
4.4.2 Effects of chronic levosimendan therapy on MI size, LV function and remodelling
Myocardial infarct size was determined by TTC-stained tissue samples and using a
reduction of 40% as a cut-off in the initial uptake rate (k1) of [11C]acetate PET.
Left ventricular function was evaluated by measurement of end-diastolic and endsystolic volumes and ejection fraction from CT images.
Cardiac remodelling was evaluated by calculation of work-metabolic index reflecting
myocardial efficiency. Myocyte hypertrophy and amount of fibrosis was estimated by
histology.
4.5 Statistical analyses
Results were expressed as means ± standard deviation. A Shapiro-Wilk test was applied
to determine whether the data were normally distributed. Correlation between study
characteristics was analysed using either Pearson or Spearman correlation. Statistical
significances between two study groups were evaluated by Mann-Whitney U test or
two-tailed t-test. Statistical significances between more than two study groups were
evaluated with one-way ANOVA and Bonferroni post hoc tests. P-values <0.05 were
considered significant.
5.
Results41
RESULTS
5.1 Atherosclerotic plaque inflammation (I)
The principal study consisted of 10 pigs with a 6-month diet intervention. Pigs were
hyperglycemic and hypercholesterlomic. Prior to imaging studies, the blood glucose
level was 12.3±4.7 mmol/L and plasma total cholesterol level was 12.7±5.1 mmol/L.
5.1.1 Ex vivo studies
In total, 33 coronary artery segments were prepared and studied. As histological grading
based on Movat pentachrome staining, seven segments were defined as a healthy vessel
wall, 16 as intimal thickening and 10 as an atheroma. Intimal thickening and atheroma
lesions contained a high density of inflammatory cells.
Increased [18F]FDG uptake was seen by autoradiography in coronary segments with
intimal thickening and atheroma. Lesion-to-normal vessel wall ratio was 1.7±0.7 times
higher in the areas of intimal thickening and 4.1±2.3 in the atheroma plaques.
The ex vivo biodistribution study showed increased [18F]FDG accumulation in atheroma
lesions compared to healthy coronary segments. Vessel-to-blood ratios of [18F]FDG
accumulation were 1.3±0.7, 2.0±1.0 and 2.6±1.2 in the segments with no plaque, intimal
thickening or atheroma lesions, respectively.
5.1.2 In vivo PET imaging
Coronary [18F]FDG accumulation was visualised by dual-gated cardiac PET images coregistered with CTA image (Figure 8). [18F]FDG uptake in the myocardium was low.
Average TBR was 1.1±0.5, 1.2±0.4 and 1.6±0.6 in the segments with no plaque, intimal
thickening and fibroatheroma, respectively. In dual-gated PET, the highest TBR was 2.7
whereas it was only 2.0 in non-gated PET.
Figure 8. Fused axial coronary CTA and dual-gated [18F]FDG PET images showing cross-section
of the proximal right coronary artery (arrow) in an animal with atheroma.
42
Results
5.2 Myocardial infarction and remodelling (II, III)
5.2.1 Myocardial perfusion and viability
Average MBF measured in the remote non-infarcted myocardium with [15O]water PET
was comparable between pigs with distal LAD ligation and proximal ameroid constrictor
and sham-operated control pigs (rest MBF: 0.97±0.32 vs. 1.24±0.40 mL/g/min, P=0.12;
stress MBF: 1.61±0.75 vs. 1.94±1.00 mL/g/min, P=0.43). Also, further calculations of
coronary flow reserve (CFR) and coronary vascular resistance (CVR) in the remote tissue
showed no statistical differences between study groups (CFR 1.68±0.63 in ameroid pigs,
1.60±0.65 in control group, P=0.80; CVR 107.1±31.5 in ameroid pigs, 93.0±29.2 mm
Hg/(mL/g/min) in control group, P=0.33) (II).
In the bottleneck stented pigs, a smaller perfusion defect (24±12% of the LV) was seen
at rest and larger during stress (53±10% of the LV). Four weeks after discontinuation of
antiplatelet medication, large perfusion defect areas were seen in both at rest and during
stress (42±13% and 54±10% of the LV, respectively) (Figure 9) (III).
REST 1W
STRESS 1W
REST 5W
REST 5W
STRESS 5W
STRESS 5W
0
20
40
60
80
100 %
Figure 9. Inducible ischaemia seen in perfusion maps obtained with [15O]water PET at 1 week
after placing a bottleneck stent into the left anterior descending coronary artery (LAD). After
stopping the antiplatelet medication, the stent has occluded and caused myocardial infarction as
seen at the 5-week time point.
Results43
Analysis of the [18F]FDG viability study performed in bottleneck stented pigs at four
weeks after discontinuation of antiplatelet medication showed that 36±3% of the LV
myocardium was non-viable and 36±18% viable whereas 28±15% was defined as
partially viable (III).
5.2.2 Left ventricular function (II)
CT imaging showed increased LV end-diastolic and end-systolic volumes in pigs having
distal ligation of the LAD and proximal ameroid constrictor compared to controls (EDV
252±84 vs. 145±17 mL, P=0.003; ESV 154±68 vs. 53±7 mL, P<0.001) (Figure 12).
Ejection fraction was reduced in ameroid pigs (40±8% vs. 63±4%, P<0.001). LV mass
normalised for body weight showed a higher LV mass to body weight index in ameroid
pigs (1.90±0.39 vs. 1.37±0.61 g/kg, P=0.02).
5.2.3 Myocardial oxidative metabolism and efficiency (II)
Analysis of the clearance rate (Kmono) of [11C]acetate showed that oxygen consumption
was reduced in the infarcted regions, but was comparable in the non-infarcted remote
myocardium of the ameroid pigs and sham-operated controls (remote Kmono 0.104±0.020
vs. 0.119±0.028 min-1, P=0.16). Global myocardial efficiency was lower in the ameroid
than sham-operated pigs (33.2±11.1 vs. 52.7±16.6 mmHg × mL × min × g-1 × 103,
P=0.005). Regional efficiency in the remote tissue was increased in ameroid pigs
(995±287 vs. 647±99 mmHg × min, P=0.004).
5.2.4 Tissue samples and histology (II)
TTC staining revealed that there was mostly a transmural infarct scar in the LV region
vascularised by the LAD. Based on analysis of [11C]acetate data, the size of MI was
29±14% (range 14%-57%) of the LV. MI size defined by [11C]acetate perfusion and TTC
staining corresponded well.
Measurement of cardiomyocyte diameter in samples from the remote non-infarcted
myocardium indicated increased myocyte hypertrophy in ameroid pigs (23.5±2.6 vs.
21.4±1.6 µm, P=0.04). The amount of fibrosis was comparable between the study groups
(5.5±1.9% in ameroid pigs; 4.9±1.1% in control group, P=0.39).
5.3 Evaluation of new perfusion tracers (IV)
5.3.1 PET imaging and kinetic modelling of [68Ga] ligands
PET imaging of four 68Ga labelled hexadentate bis(salicylaldimine) ligands showed the
highest and fastest uptake into myocardium on Tracer-3 (Figure 10).
44
46
Results
Results
Tracer-1
Tracer-2
Tracer-3
Tracer-4
Figure
10. Representative horizontal long-axis view of heart showing the myocardial uptake of
Figure
10. Representative horizontal long-axis view of heart showing the myocardial uptake of the
68
the
Ga tracers at 52-92 min after the tracer injection.
68
Ga tracers at 52-92 min after the tracer injection.
one-tissue
compartmental
model model
(K1) were
ModellingModelling
results were
Patlak plots
i) and
Patlak
plots(K(K
) and
one-tissue
compartmental
(K1calculated.
) were calculated.
i
15
15
There was
no correlation
between
and MBF
plotted against
results
obtained
with results
[ O]water.
ThereKiwas
no
results
were MBF
plotted
against
MBF
obtained
with
[ O]water.
15
15
O]water.
There
tended
to
be
a
weak
correlation
between
K
and
MBF
for
Tracer-1
measured
with
[
1
correlation between Ki and MBF measured with [ O]water. There tended to be a weak
(r=0.81, P=0.20),
but not
anyMBF
other for
tracer.
and
Tracer-1 (r=0.81, P=0.20), but not for any other
correlation
between
K1for
tracer.
5.3.2 Organ distribution
5.3.2
distribution
labelled hexadentate bis(salicylaldimine) ligands was slightly higher in the
UptakeOrgan
of 68Ga
myocardium
in other examined
organs.
Accumulation in kidney
andwas
bile slightly
were highhigher
indicating
Ga labelled
hexadentate
bis(salicylaldimine)
ligands
in
Uptake
of 68than
elimination and excretion routes. Tracer-3 showed good myocardium-to-blood (7.63±1.89),
the myocardium than in other examined organs. Accumulation in kidney and bile were
myocardium-to-lung (3.03±0.33) and myocardium-to-liver (1.80±0.82) ratios.
high indicating elimination and excretion routes. Tracer-3 showed good myocardiumto-blood (7.63±1.89), myocardium-to-lung (3.03±0.33) and myocardium-to-liver
(1.80±0.82) ratios.
5.3.3
In vitro binding to serum proteins
5.3.3 In vitro binding to serum proteins
A high difference in tracer binding to serum proteins was seen between human, pig and rat serum.
Thehigh
unbound
tracer fraction
was
similarly
between
human
serum
pig serum
A
difference
in tracer
binding
to low
serum
proteins
was
seen(9.4±5.7%)
between and
human,
pig
(5.5±2.1%),
whereas
was markedly
higher
in rat serum
(45.4±20.9%).
and
rat serum.
Theit unbound
tracer
fraction
was similarly
low between human serum
(9.4±5.7%) and pig serum (5.5±2.1%), whereas it was markedly higher in rat serum
(45.4±20.9%).
5.4
5.4.1
Evaluation of chronic levosimendan intervention for heart failure (V)
Effects of chronic levosimendan therapy on MI size, LV function and remodelling
5.4 Evaluation of chronic levosimendan intervention for heart failure (V)
5.4.1
Effectsafter
of chronic
levosimendan
therapy
on MI size,
LV function
remodelling
As measured
the 8-week
intervention period
following
one week
washout,and
myocardial
infarct
size measured
was smallerafter
in thethe
levosimendan
group when compared
with control
vs.
As
8-week intervention
period following
oneanimals
week(12±13%
washout,
27±15%, P=0.03).
myocardial infarct size was smaller in the levosimendan group when compared with
control
animals
(12±13%
CT imaging
showed
smaller vs.
LV27±15%,
EDV and P=0.03).
ESV in the levosimendan group (EDV 161±29 mL vs.
245±84 mL, P = 0.06; ESV 81±18 mL vs. 149±67 mL, P=0.03). The ejection fraction tended to be
CT
imaging
showed smaller
LV(50±6%
EDV and
ESV inP=0.06).
the levosimendan group (EDV 161±29
higher
in the levosimendan
group
vs. 41±8%,
mL vs. 245±84 mL, P = 0.06; ESV 81±18 mL vs. 149±67 mL, P=0.03). The ejection
fraction tended to be higher in the levosimendan group (50±6% vs. 41±8%, P=0.06).
Results45
Myocardial efficiency remained unchanged after intervention and was comparable
between study groups (44±11 in the levosimendan group and 33±11 mmHg × mL × min
× g-1 × 103 in the control group, P=0.15).
Histological analysis of non-infarcted remote sections indicated that the average diameter
of cardiomyocytes (levosimendan group 23.2±2.0 µm vs. control group 23.5±2.6 µm,
P=0.83) and the amount of fibrosis (4.8±1.9% in the levosimendan group; 5.5±1.9% in
the control group, P=0.37) were comparable between study groups.
46
6.
Discussion DISCUSSION
Non-invasive assessment of atherosclerotic inflammation can be beneficial to recognise
patients having a high risk for cardiac events. Early detection of highly inflamed
atherosclerotic lesions could give us an opportunity to identify unstable plaques, improve
the care of CAD patients and reduce the risk of sudden cardiac death.
Myocardial infarction often leads to adverse cardiac remodelling and finally heart failure.
Oxidative metabolism and perfusion of myocardial tissue can be evaluated by [11C]
acetate PET. Additionally, efficiency of cardiac work can be calculated. In this study we
wanted to validate the HF model of distal LAD ligation and proximal ameroid constrictor
with the evaluation of perfusion, oxidative metabolism and cardiac efficiency.
As the prognosis of heart failure is poor, new therapies for the management of progression
of post-MI adverse cardiac remodelling and heart failure are warranted. As earlier studies
have already shown levosimendan could have multiple positive effects to prevent adverse
cardiac remodelling. This study proved the beneficial effects of levosimendan as well as
that the developed model of HF can be used for the evaluation of new interventions.
6.1 PET imaging of early atherosclerotic lesions
Imaging of atherosclerotic inflammation by [18F]FDG PET could be valuable for
identifying high-risk patients. Increased [18F]FDG accumulation exists in culprit lesions
by in vivo imaging studies (Rogers et al. 2010). In this study we evaluated [18F]FDG
uptake in early atherosclerotic lesions by in vivo imaging and ex vivo studies. The main
finding of this study was that [18F]FDG accumulation was increased in coronary arteries
with intimal thickening and atheroma as confirmed by autoradiography and histological
evaluation.
PET imaging of coronary arteries may be an effective method to differentiate
atherosclerosis patients having highly inflamed vulnerable plaques or stable plaques
with low inflammatory activity. The dual-gated method for minimising the cardiac and
respiratory movement artefact increased TBR as the highest TBR in dual-gated PET was
2.7 and 2.0 in non-gated PET that is consistent with previous studies (Tarkin et al. 2014).
Still, it was not statistically possible to differentiate [18F]FDG uptake between a healthy
vessel wall or one with intimal thickening or an atheroma. The sensitivity of in vivo PET
is probably not sensitive enough for detecting small objects with a low accumulation of
imaging tracer.
Advanced atherosclerotic lesions with coronary stenosis was reported in a previous study
with a similar study setting that combined hyperglycemia and hypercholesterolemia
(Gerrity et al. 2001 and Chatzizisis et al. 2008). However, very early atherosclerotic
Discussion 47
changes were noticed in the current study. Coronary stenosis was not identified as
confirmed with CTA and histology.
Further studies are warranted to clarify if more advanced atherosclerotic lesions can
be induced within a reasonable follow-up time. PET imaging of atherosclerotic lesions
showed to be effective and the dual-gating method improved the signal-to-noise ratio.
Clinically relevant CAD model could be beneficial in developing further clinical
diagnostics and treatments.
6.2 Validation of a surgically induced model of myocardial infarction
A translational research model to study chronic changes after MI is needed. In this study,
cardiac remodelling was followed 3 months after MI. Two-step occlusion of LAD with
distal ligation and proximal ameroid constrictor led to MI covering 24 percent of the LV
as confirmed with [11C]acetate perfusion imaging and TTC staining of myocardial organ
samples.
LV size was increased over 70 percent whereas EF was decreased 37 percent. Global
myocardial efficiency was decreased 37 percent, which is connected to HF. Increased
LV size led to increased myocardial wall stress. Histologically-detectable myocyte
hypertrophy is linked to cardiac remodelling and is a compensatory mechanism
for increased wall stress. Interestingly, interstitial fibrosis was not seen as described
previously in a similar study (Teramoto et al. 2011).
As high as a 75 percent survival rate was reported by the authors of the original model
description (Teramoto et al. 2011). In this study, the overall survival was only 26%. Still,
most of unexpected sudden deaths occurred during the first week of the surgery and
chronic phase survival was 44%. Sudden cardiac deaths are common in other pig studies
and are probably linked to large MI size (Fallavollita et al. 2005, Huang et al. 2010 and
Ishikawa et al. 2011).
In this study, we confirmed the feasibility of a chronic post-MI heart failure model
utilising two-step occlusion of the LAD with distal ligation followed by implantation
of an ameroid constrictor in the proximal part. Left ventricular hypertrophy, impaired
systolic function and LV remodelling were confirmed by imaging studies and histological
evaluation. Measurements of myocardial oxidative metabolism demonstrated reduced
cardiac efficiency. We conclude that this model may be used for the evaluation of new
interventions.
As the mortality observed in this study was very high, further development of the model
of chronic HF is needed. This study furthered the understanding behind the mechanisms
of cardiac remodelling. Further studies relating to ischaemic preconditioning and
preventing adverse cardiac remodelling post-MI are needed.
48
Discussion 6.3 Validation of percutaneously-induced model of myocardial ischaemia
and infarction
Bottleneck stenting of LAD induced large ischaemic region as measured with [15O]water
(24% of LV at rest and 53% of LV during stress). Discontinuing of antiplatelet medication
led to an increased defect area (42% of LV at rest and 54% of LV during stress).
As measured by [18F]FDG, the non-viable portion of LV was 36 percent at four weeks
after discontinuing of antiplatelet medication. Infarct size was large referring to the
clinical situation and this measurement makes it possible to evaluate processes after
MI.
An advantage of this model compared to ameroid model is that chronic myocardial
ischaemia can be studied by continuing antiplatelet medication. Also, a lack of adhesion
and inflammation induced by wound healing occurring after thoracic surgery noticed
in surgical MI model can be beneficial in studies based on surgical interventions.
Inflammation induced by wound healing can influence negatively also imaging studies
of inflammation.
6.4 PET imaging and kinetic modelling of [68Ga] ligands
Studied 68Ga-labelled hexadentate bis(salicylaldimine) ligands were retained in the
myocardium as shown in previous study with rats (Hsiao et al. 2009). Uptake into the
myocardium was slow as well as clearance from blood pool. Kinetic modelling showed
that myocardial perfusion cannot be determined with these four [68Ga] ligands as
compared to MBF results obtained with [15O]water.
There was no significant correlation between results obtained with [15O]water and the net
uptake rate Ki (Patlak plot) and K1 (one-compartmental model). One possible explanation
for slow kinetics could be that tracers are very slowly moved across the cell membranes.
A slow transport rate is a limiting factor for Ki and K1. An unknown factor other than
perfusion may be limiting the accumulation into the myocardium. Another explanation
could be a high tracer binding to serum proteins (91%-96% in pig vs. 29%-73% in rat)
because the protein-bound fraction tracers are not expected to freely diffuse into tissues,
which is one requirement for an effective perfusion tracer.
A slow transport rate and high protein binding together could be the explanation for the
lack of correlation between MBF measured with [15O]water and [68Ga] ligands as tested
in healthy pigs.
Despite the lack of correlation between tested [68Ga] tracers and [15O]water in this study
with pigs, further studies are needed to clarify the mechanisms of the [68Ga] ligands,
which showed to be promising for MPI in a previous rat study.
Discussion 49
6.5 Chronic levosimendan intervention for heart failure
In this study, the effects of 8-week levosimendan intervention, started three weeks after
distal ligation of the LAD and placement of proximal ameroid constrictor, were studied.
A significant reduction in MI size was seen in animals that received levosimendan 5
mg/kg for 8 weeks. Previously this kind of cardioprotection and reduced MI sizes were
reported in a study where levosimendan therapy was initiated before inducing myocardial
ischaemia and infarction (Kersten et al. 2000). In this study, a LV wall motion defect
was confirmed by echocardiography before allocating animals into study groups and
beginning the therapy.
Animals in the levosimendan group have also smaller EDV and ESV and higher EF than
animals in the control group. That is probably explained by a smaller MI size. These
results are consistent with the results of previous small animal studies.
Beneficial effects of levosimendan in this study are possibly explained by mitochondrial
KATP channel-mediated preconditioning, which protects myocytes against ischaemia
(Gross et al. 1992). Also, increased collateral blood flow has been noticed after
levosimendan administration (Kersten et al. 2000).
Levosimendan intervention restricted the size of MI and further cardiac remodelling, but
more studies are needed to explain the exact mechanisms. Levosimendan is used for the
management of acutely decompensated heart failure. Based on our results, levosimendan
could have benefits also in the early treatment of acute myocardial infarction. Large
animal models and clinical imaging methods used in this study enable quantitative
analysis of myocardial perfusion and efficiency. This study that experimental pig model
for CAD, MI, and HF could be used for further intervention studies.
6.6 Critical evaluation of the results
Large animal models of atherosclerosis enable the evaluation of coronary arteries by
using in vivo imaging methods. In this study we were able to induce early atherosclerotic
changes including intimal thickening and atheroma-type lesions in relation to clinical
coronary artery disease. This study showed how ex vivo methods can be used in a
validation of in vivo results obtained by clinical imaging device. Despite the relatively
long follow-up time we did not noticed any narrowing of coronary arteries with
anatomical imaging methods. Also, highly inflamed coronary lesions clinically related
to increased risk for cardiac events were lacking. Long follow-up led to high expenses
due to costly animal housing and special high-fat diet. Also, a significant increase in
body mass in short periods of time led to difficulties in animal handling.
Surgical protocol consisted of distal ligation of the LAD and proximal ameroid constrictor
led to large myocardial infarction following LV adverse cardiac remodelling in relation
to clinical picture of myocardial infarction and heart failure. Even though we used distal
50
Discussion ligation of the LAD to induce ischaemic preconditioning, a high amount of sudden deaths
were noticed in this study. Intolerance of myocardial ischaemia leading to high mortality
was one important limitation in this study. Mortality in this study was higher than in
HF patients and is probably not reflecting well the clinical picture. Additionally, in the
clinic, HF is mainly a problem in older adults. Fairly young animals need to be used in
experiments. Physiologic increases in heart mass during follow-up can be avoided by
using models involving adult minipigs (Schuleri et al. 2008).
A bottleneck stent placed into the LAD led to the situation mimicking clinically relevant
myocardial ischaemia and infarction. Premature occlusion of coronary artery was
prevented by antiplatelet medication, which can lead to bleeding complications. Also, a
high quality coronary angiography device and good operator skills are needed to perform
this percutaneous method.
The evaluation of myocardial perfusion with 68Ga-labelled hexadentate bis(salicylaldimine)
ligands was positive in rat studies. In this study with healthy pigs, these ligands were
retained into myocardium, but the kinetics were very slow. We noticed that tracers
were binding highly to serum proteins. Binding properties to serum proteins were very
similar between pig and human (91%-96% in pig vs. 83%-96% in human), whereas
it was remarkable different in rat (29%-73%). This large animal model was better for
translational studies because it is more close to human metabolism than rodent models
in this case.
Chronic levosimendan therapy tested in chronic MI model showed to have beneficial
properties to restrict the size of MI and cardiac remodelling post-MI. Limitations in this
study are related to high variability in the size of MI and imbalance of study population
between control and intervention group.
6.7 Future aspects
Experimental pig models of atherosclerosis having myocardial ischaemia and
infarction offer a versatile platform for translational research. In the future, more
novel radiopharmaceuticals and interventions can be studied. Novel therapeutics
can be focused on prevention of atherosclerotic plaque formation and rupture. New
interventions concerning heart failure could consist of an innovative batch materials
replacing infarcted myocardium. Also, gene and stem cell therapies are promising to
improve cardiac function after MI. Large animal models with a human-like clinical
picture are effective and valuable tools for biomedical research.
Large animal models are needed because it is not possible to study complex mechanisms
of atherosclerosis and HF with alternative methods like cell cultures. The size of pig
enables the imaging studies of coronary arteries. In small animal models, we need to use
another vessel like the aorta for modelling of atherosclerosis. In large animal studies,
we can use already existing methods and devices dedicated to clinical studies and thus
Discussion 51
new innovations are more easily translated into clinical use. Despite many advantages
of large animal models, there are also some disadvantages, which might be taken into
account in study planning like difficulties in handling of heavy animals. Large animal
studies need special facilities for animal housing and operations and can lead to high
expenses. The use of large animal models remains restricted to research of new concepts
close to clinical application.
More advanced mechanisms behind atherosclerosis and heart failure can be evaluated
with multimodality imaging methods. We and others have evaluated the imaging of
different stages of atherosclerotic changes with [18F]FDG PET, but it is still unsolved
how to use this information in clinical imaging of atherosclerotic patients in the future.
Myocardial oxidative metabolism and further assessment of cardiac efficiency is possible
with [11C]acetate PET. Still, these methods are relatively rarely used in clinical imaging
and thus have a huge potential in monitoring of efficacy of treatment in HF patients.
Large animals enable the use of clinical imaging methods and thus new interventions can
be studied in translational models before entering clinical trials.
52
7.
Summary and Conclusions SUMMARY AND CONCLUSIONS
On the basis of our experimental studies on atherosclerosis and heart failure, we make
these conclusions:
1.
Model of diabetes and hypercholesterolemia was feasible and we found an
increased uptake of [18F]FDG in early coronary atherosclerotic lesions in a pig
model of diabetes and hypercholesterolemia.
2.
Two-step occlusion of the LAD with distal ligation and proximal ameroid
constrictor resulted in a large MI and LV remodelling together with decreased
myocardial efficiency during a 3-month follow-up.
3.
The bottleneck stent placed in the LAD resulted in a large ischaemic region of
myocardium followed by large MI after discontinuing of antiplatelet medication.
4.
Evaluation of four 68Ga-labelled hexadentate bis(salicylaldimine) ligands showed
no correlation between myocardial perfusion measured with [15O]water.
5.
Eight-week levosimendan therapy started after recent occlusion of the LAD
resulted in decreased MI size followed by attenuation of myocardial remodelling
and improved systolic function.
Large animal models mimicking clinical conditions and multimodality imaging can be
used as valuable tools in translational research of atherosclerosis and HF. Large animals
enable the use of clinical imaging methods and thus new interventions can be studied in
translational models before entering clinical trials.
9.
References55
REFERENCES
Aarsæther, E., Straumbotn, E., Rösner, A. and Busund,
R. 2012. “Oral Β-Glucan Reduces Infarction Size
and Improves Regional Contractile Function in a
Porcine Ischaemia/reperfusion Model.” European
Journal of Cardio-Thoracic Surgery : Official
Journal of the European Association for CardioThoracic Surgery 41 (4): 919–25.
Abdel-Rahman, U., Risteski, P., Tizi, K., Kerscher,
S., Behjati, S., Bejati, S., Zwicker, K., Scholz,
M., Brandt, U. and Moritz, A. 2009. “Hypoxic
Reoxygenation
during
Initial
Reperfusion
Attenuates Cardiac Dysfunction and Limits
Ischemia-Reperfusion Injury after Cardioplegic
Arrest in a Porcine Model.” The Journal of Thoracic
and Cardiovascular Surgery 137 (4): 978–82.
Abegunewardene, N., Vosseler, M., Gori, T.,
Hoffmann, N., Schmidt, K.-H., Becker, D.,
Kreitner, K.-F. et al. 2009. “Usefulness of MRI to
Differentiate between Temporary and Long-Term
Coronary Artery Occlusion in a Minimally Invasive
Model of Experimental Myocardial Infarction.”
Cardiovascular and Interventional Radiology 32
(5): 1033–41.
Aliev, G. and Burnstock, G. 1998. “Watanabe Rabbits
with Heritable Hypercholesterolaemia: A Model of
Atherosclerosis.” Histology and Histopathology 13
(3): 797–817.
Allman, K.C. 2013. “Noninvasive Assessment
Myocardial Viability: Current Status and Future
Directions.” Journal of Nuclear Cardiology 
:
Official Publication of the American Society of
Nuclear Cardiology 20 (4): 618–37; quiz 638–39.
Al-Mashhadi, R.H., Sørensen, C.B., Kragh, P.M.,
Christoffersen, C., Mortensen, M.B., Tolbod, L.P.,
Thim, T. et al. 2013. “Familial Hypercholesterolemia
and Atherosclerosis in Cloned Minipigs Created by
DNA Transposition of a Human PCSK9 Gain-ofFunction Mutant.” Science Translational Medicine
5 (166): 166ra1.
Angeli, F.S., Amabile, N., Burjonroppa, S., Shapiro,
M., Bartlett, L., Zhang, Y., Virmani, R. et al. 2010.
“Prolonged Therapy with Erythropoietin Is Safe
and Prevents Deterioration of Left Ventricular
Systolic Function in a Porcine Model of Myocardial
Infarction.” Journal of Cardiac Failure 16 (7): 579–
89.
Angeli, F.S., Amabile, N., Shapiro, M., Mirsky, R.,
Bartlett, L., Zhang, Y., Virmani, R. et al. 2010.
“Cytokine Combination Therapy with Erythropoietin
and Granulocyte Colony Stimulating Factor in a
Porcine Model of Acute Myocardial Infarction.”
Cardiovascular Drugs and Therapy / Sponsored
by the International Society of Cardiovascular
Pharmacotherapy 24 (5-6): 409–20.
Antonov, A.S., Kolodgie, F.D., Munn, D.H. and Gerrity,
R.G. 2004. “Regulation of Macrophage Foam Cell
Formation by alphaVbeta3 Integrin: Potential Role
in Human Atherosclerosis.” The American Journal
of Pathology 165 (1): 247–58.
Armbrecht, J.J., Buxton, D.B. and Schelbert, H.R.
1990. “Validation of [1-11C]acetate as a Tracer for
Noninvasive Assessment of Oxidative Metabolism
with Positron Emission Tomography in Normal,
Ischemic, Postischemic, and Hyperemic Canine
Myocardium.” Circulation 81: 1594–1605.
Arslan, F., Houtgraaf, J.H., Keogh, B., Kazemi, K.,
de Jong, R., McCormack, W.J., O’Neill, L.A.J. et
al. 2012. “Treatment with OPN-305, a Humanized
Anti-Toll-Like Receptor-2 Antibody, Reduces
Myocardial Ischemia/reperfusion Injury in Pigs.”
Circulation. Cardiovascular Interventions 5 (2):
279–87.
Artinger, S., Deiner, C., Loddenkemper, C.,
Schwimmbeck, P.L., Schultheiss, H.-P. and Pels, K.
2009. “Complex Porcine Model of Atherosclerosis:
Induction of Early Coronary Lesions after Long-Term
Hyperlipidemia without Sustained Hyperglycemia.”
The Canadian Journal of Cardiology 25 (4): e109–
14.
Ayala-López, W., Xia, W., Varghese, B. and Low, P.S.
2010. “Imaging of Atherosclerosis in Apoliprotein E
Knockout Mice: Targeting of a Folate-Conjugated
Radiopharmaceutical to Activated Macrophages.”
Journal of Nuclear Medicine : Official Publication,
Society of Nuclear Medicine 51 (5): 768–74.
Bahls, M., Bidwell, C.A., Hu, J., Krueger, C.G.,
Reed, J.D., Tellez, A., Kaluza, G.L., Granada,
J.F., Van Alstine, W.G. and Newcomer, S.C.
2011. “Gene Expression Differences in Healthy
Brachial and Femoral Arteries of Rapacz Familial
Hypercholesterolemic
Swine.”
Physiological
Genomics 43 (12): 781–88.
Bamberg, F., Hinkel, R., Schwarz, F., Sandner, T.A.,
Baloch, E., Marcus, R., Becker, A. et al. 2012.
“Accuracy of Dynamic Computed Tomography
Adenosine Stress Myocardial Perfusion Imaging
in Estimating Myocardial Blood Flow at Various
Degrees of Coronary Artery Stenosis Using a
Porcine Animal Model.” Investigative Radiology 47
(1): 71–77.
56
References
Banz, Y., Rieben, R., Zobrist, C., Meier, P., Shaw,
S., Lanz, J., Carrel, T. and Berdat, P. 2008.
“Addition of Dextran Sulfate to Blood Cardioplegia
Attenuates Reperfusion Injury in a Porcine Model
of Cardiopulmonary Bypass.” European Journal of
Cardio-Thoracic Surgery : Official Journal of the
European Association for Cardio-Thoracic Surgery
34 (3): 653–60.
Bertsch, T., Janke, C., Denz, C., Weiss, M., Luiz,
T., Ellinger, K., Korth, U., Hannak, D., Bartelt,
U. and Krieter, H. 2000. “Cardiac Troponin I
and Cardiac Troponin T Increases in Pigs during
Ischemia-Reperfusion Damage.” Experimental
and Toxicologic Pathology : Official Journal of the
Gesellschaft Für Toxikologische Pathologie 52 (2):
157–59.
Barallobre-Barreiro, J., Didangelos, A., Schoendube,
F.A., Drozdov, I., Yin, X., Fernández-Caggiano,
M., Willeit, P. et al. 2012. “Proteomics Analysis
of Cardiac Extracellular Matrix Remodeling in a
Porcine Model of Ischemia/reperfusion Injury.”
Circulation 125 (6): 789–802.
Bhindi, R., Fahmy, R.G., McMahon, A.C., Khachigian,
L.M. and Lowe, H.C. 2012. “Intracoronary Delivery
of DNAzymes Targeting Human EGR-1 Reduces
Infarct Size Following Myocardial Ischaemia
Reperfusion.” The Journal of Pathology 227 (2):
157–64.
Barandon, L., Calderon, J., Réant, P., Caillaud, D.,
Lafitte, S., Roques, X., Couffinhal, T. and Dos
Santos, P. 2010. “Adjustment and Characterization
of an Original Model of Chronic Ischemic Heart
Failure in Pig.” Cardiology Research and Practice
2010.
Bigalke, B., Phinikaridou, A., Andia, M.E., Cooper,
M.S., Schuster, A., Wurster, T., Onthank, D. et
al. 2014. “PET/CT and MR Imaging Biomarker
of Lipid-Rich Plaques Using [64Cu]-Labeled
Scavenger Receptor (CD68-Fc).” International
Journal of Cardiology 177 (1): 287–91.
Basken, N.E., Mathias, C.J., Lipka, A.E. and Green,
M.A. 2008. “Species Dependence of [64Cu]CuBis(thiosemicarbazone)
Radiopharmaceutical
Binding to Serum Albumins.” Nuclear Medicine
and Biology 35 (3): 281–86.
Biondi-Zoccai, G., De Falco, E., Peruzzi, M.,
Cavarretta, E., Mancone, M., Leoni, O., Caristo,
M.E. et al. 2013. “A Novel Closed-Chest Porcine
Model of Chronic Ischemic Heart Failure Suitable
for Experimental Research in Cardiovascular
Disease.” BioMed Research International 2013
(January): 410631.
Beer, A.J., Pelisek, J., Heider, P., Saraste, A., Reeps,
C., Metz, S., Seidl, S. et al. 2014. “PET/CT Imaging
of Integrin αvβ3 Expression in Human Carotid
Atherosclerosis.” JACC. Cardiovascular Imaging 7
(2): 178–87.
Bengel, F.M., Permanetter, B., Ungerer, M., Nekolla, S.
and Schwaiger, M. 2000. “Non-Invasive Estimation
of Myocardial Efficiency Using Positron Emission
Tomography and Carbon-11 Acetate--Comparison
between the Normal and Failing Human Heart.”
Eur.J.Nucl.Med. 27: 319–26.
Bergmann, S.R., Fox, K.A., Rand, A.L., McElvany,
K.D., Welch, M.J., Markham, J. and Sobel, B.E.
1984. “Quantification of Regional Myocardial
Blood Flow in Vivo with H215O.” Circulation 70
(4): 724–33.
Bertho, E. and Gagnon, G. 1964. “A Comparative
Study in Three Dimension of the Blood Supply
of the Normal Interventricular Septum in Human,
Canine, Bovine, Procine, Ovine and Equine Heart.”
Diseases of the Chest 46 (September): 251–62.
Bertsch, T., Denz, C., Janke, C., Weiss, M., Fassbender,
K., Luiz, T., Ellinger, K. and Krieter, H. 2001.
“Hypertonic-Hyperoncotic Solutions Decrease
Cardiac Troponin I Concentrations in Peripheral
Blood in a Porcine Ischemia-Reperfusion Model.”
Experimental and Toxicologic Pathology : Official
Journal of the Gesellschaft Für Toxikologische
Pathologie 53 (2-3): 153–56.
Bisplinghoff, S., Hänisch, C., Becker, M., Radermacher,
K. and de la Fuente, M. 2014. “Fusion of Coronary
Angiography and Stress Echocardiography for
Myocardial Viability Evaluation.” International
Journal of Computer Assisted Radiology and
Surgery, May.
Bito, V., Heinzel, F.R., Weidemann, F., Dommke, C.,
van der Velden, J., Verbeken, E., Claus, P. et al. 2004.
“Cellular Mechanisms of Contractile Dysfunction in
Hibernating Myocardium.” Circulation Research 94
(6): 794–801.
Bloor, C.M., White, F.C. and Sanders, T.M. 1984.
“Effects of Exercise on Collateral Development in
Myocardial Ischemia in Pigs.” Journal of Applied
Physiology: Respiratory, Environmental and
Exercise Physiology 56 (3): 656–65.
Boekstegers, P., Raake, P., Al Ghobainy, R., Horstkotte,
J., Hinkel, R., Sandner, T., Wichels, R. et al. 2002.
“Stent-Based Approach for Ventricle-to-Coronary
Artery Bypass.” Circulation 106 (8): 1000–1006.
Bolukoglu, H., Liedtke, A.J., Nellis, S.H., Eggleston,
A.M., Subramanian, R. and Renstrom, B. 1992.
“An Animal Model of Chronic Coronary Stenosis
Resulting in Hibernating Myocardium.” Am J
Physiol Heart Circ Physiol 263 (1): H20–29.
References57
Brødløs, H.K., Bramsen, M.B., Agger, P., Jensen, H.,
Bjerre, M., Ringgaard, S., Wierup, P., Nielsen, S.L.,
Hasenkam, J.M. and Smerup, M. 2009. “A Catheter
Based Chronic Porcine Model of Post-Infarct Dilated
Heart Failure.” Scandinavian Cardiovascular
Journal : SCJ 43 (4): 260–66.
Buja, L.M. 2005. “Myocardial Ischemia and
Reperfusion Injury.” Cardiovascular Pathology :
The Official Journal of the Society for Cardiovascular
Pathology 14 (4): 170–75.
Bujak, M. and Frangogiannis, N.G. 2007. “The Role
of TGF-Beta Signaling in Myocardial Infarction and
Cardiac Remodeling.” Cardiovascular Research 74
(2): 184–95.
Caillaud, D., Calderon, J., Réant, P., Lafitte, S., Dos
Santos, P., Couffinhal, T., Roques, X. and Barandon,
L. 2010. “Echocardiographic Analysis with a TwoDimensional Strain of Chronic Myocardial Ischemia
Induced with Ameroid Constrictor in the Pig.”
Interactive Cardiovascular and Thoracic Surgery
10 (5): 689–93.
Carter, A.J., Laird, J.R., Kufs, W.M., Bailey, L.,
Hoopes, T.G., Reeves, T., Farb, A. and Virmani, R.
1996. “Coronary Stenting with a Novel Stainless
Steel Balloon-Expandable Stent: Determinants
of Neointimal Formation and Changes in Arterial
Geometry after Placement in an Atherosclerotic
Model.” Journal of the American College of
Cardiology 27 (5): 1270–77.
Cason, B.A., Demas, K.A., Mazer, C.D., Gordon, H.J.
and Hickey, R.F. 1991. “Effects of Nitrous Oxide
on Coronary Pressure and Regional Contractile
Function in Experimental Myocardial Ischemia.”
Anesthesia and Analgesia 72 (5): 604–11.
Cerqueira, M.D., Weissman, N.J., Dilsizian, V.,
Jacobs, A.K., Kaul, S., Laskey, W.K., Pennell,
D.J., Rumberger, J.A., Ryan, T. and Verani, M.S.
2002. “Standardized Myocardial Segmentation and
Nomenclature for Tomographic Imaging of the
Heart. A Statement for Healthcare Professionals
from the Cardiac Imaging Committee of the Council
on Clinical Cardiology of the American Heart
Association.” Circulation 105: 539–42.
Chatziathanasiou, G.N., Nikas, D.N., Katsouras,
C.S., Kazakos, N.D., Bouba, V., Vougiouklakis, T.,
Naka, K.K. and Michalis, L.K. 2012. “Combined
Intravenous Treatment with Ascorbic Acid and
Desferrioxamine to Reduce Myocardial Reperfusion
Injury in an Experimental Model Resembling the
Clinical Setting of Primary PCI.” Hellenic Journal
of Cardiology : HJC = Hellēnikē Kardiologikē
Epitheōrēsē 53 (3): 195–204.
Chatzizisis, Y.S., Jonas, M., Beigel, R., Coskun,
A.U., Baker, A.B., Stone, B. V, Maynard, C. et al.
2009. “Attenuation of Inflammation and Expansive
Remodeling by Valsartan Alone or in Combination
with Simvastatin in High-Risk Coronary
Atherosclerotic Plaques.” Atherosclerosis 203 (2):
387–94.
Chatzizisis, Y.S., Jonas, M., Coskun, A.U., Beigel,
R., Stone, B. V, Maynard, C., Gerrity, R.G. et al.
2008. “Prediction of the Localization of High-Risk
Coronary Atherosclerotic Plaques on the Basis of
Low Endothelial Shear Stress: An Intravascular
Ultrasound and Histopathology Natural History
Study.” Circulation 117 (8): 993–1002.
Chen, C., Chen, L., Fallon, J.T., Ma, L., Li, L., Bow,
L., Knibbs, D., McKay, R., Gillam, L.D. and Waters,
D.D. 1996. “Functional and Structural Alterations
With 24-Hour Myocardial Hibernation and Recovery
After Reperfusion: A Pig Model of Myocardial
Hibernation.” Circulation 94 (3): 507–16.
Chen, C., Ma, L., Dyckman, W., Santos, F., Lai,
T., Gillam, L.D. and Waters, D.D. 1997. “Left
Ventricular Remodeling in Myocardial Hibernation.”
Circulation 96 (9 Suppl): II – 46–50.
Chen, C., Ma, L., Linfert, D.R., Lai, T., Fallon, J.T.,
Gillam, L.D., Waters, D.D. and Tsongalis, G.J.
1997. “Myocardial Cell Death and Apoptosis in
Hibernating Myocardium.” Journal of the American
College of Cardiology 30 (5): 1407–12.
Chen, Y., Shao, D.-B., Zhang, F.-X., Zhang, J., Yuan, W.,
Man, Y.-L., Du, W. et al. 2013. “Establishment and
Evaluation of a Swine Model of Acute Myocardial
Infarction and Reperfusion-Ventricular FibrillationCardiac Arrest Using the Interventional Technique.”
Journal of the Chinese Medical Association : JCMA
76 (9): 491–96.
Chinda, K., Palee, S., Surinkaew, S., Phornphutkul,
M., Chattipakorn, S. and Chattipakorn, N. 2013.
“Cardioprotective Effect of Dipeptidyl Peptidase-4
Inhibitor during Ischemia-Reperfusion Injury.”
International Journal of Cardiology 167 (2): 451–
57.
Cho, K.R., Choi, J.-S., Hahn, W., Kim, D.S., Park,
J.S., Lee, D.S. and Kim, K.-B. 2008. “Therapeutic
Angiogenesis Using Naked DNA Expressing
Two Isoforms of the Hepatocyte Growth Factor
in a Porcine Acute Myocardial Infarction Model.”
European Journal of Cardio-Thoracic Surgery :
Official Journal of the European Association for
Cardio-Thoracic Surgery 34 (4): 857–63.
Choy, J.S., Zhang, Z.-D., Pitsillides, K., Sosa, M. and
Kassab, G.S. 2014. “Longitudinal Hemodynamic
Measurements in Swine Heart Failure Using a Fully
Implantable Telemetry System.” PloS One 9 (8):
e103331.
58
References
Christian, T.F., Peters, K., Keck, B., Allen, J., Owens,
T. and Borah, B. 2008. “Gated SPECT Imaging to
Detect Changes in Myocardial Blood Flow during
Progressive Coronary Occlusion.” The International
Journal of Cardiovascular Imaging 24 (3): 269–76.
Crick, S.J., Sheppard, M.N., Ho, S.Y., Gebstein, L.
and Anderson, R.H. 1998. “Anatomy of the Pig
Heart: Comparisons with Normal Human Cardiac
Structure.” Journal of Anatomy 193 ( Pt 1 (July):
105–19.
Crisóstomo, V., Maestre, J., Maynar, M., Sun, F.,
Báez-Díaz, C., Usón, J. and Sánchez-Margallo, F.M.
2013. “Development of a Closed Chest Model of
Chronic Myocardial Infarction in Swine: Magnetic
Resonance Imaging and Pathological Evaluation.”
ISRN Cardiology 2013 (January): 781762.
Cui, J., Li, J., Mathison, M., Tondato, F., Mulkey,
S.P., Micko, C., Chronos, N.A.F. and Robinson,
K.A. 2005. “A Clinically Relevant Large-Animal
Model for Evaluation of Tissue-Engineered
Cardiac Surgical Patch Materials.” Cardiovascular
Revascularization Medicine : Including Molecular
Interventions 6 (3): 113–20.
Dahlöf, B. 2010. “Cardiovascular Disease Risk
Factors: Epidemiology and Risk Assessment.” The
American Journal of Cardiology 105 (1 Suppl): 3A
– 9A.
Danad, I., Uusitalo, V., Kero, T., Saraste, A.,
Raijmakers, P.G., Lammertsma, A.A., Heymans,
M.W. et al. 2014. “Quantitative Assessment of
Myocardial Perfusion in the Detection of Significant
Coronary Artery Disease: Cutoff Values and
Diagnostic Accuracy of Quantitative [(15)O]H2O
PET Imaging.” Journal of the American College of
Cardiology 64 (14): 1464–75.
Das, M. and Das, D.K. 2008. “Molecular Mechanism
of Preconditioning.” IUBMB Life 60 (4): 199–203.
Dash, R., Chung, J., Ikeno, F., Hahn-Windgassen,
A., Matsuura, Y., Bennett, M. V, Lyons, J.K. et al.
2011. “Dual Manganese-Enhanced and Delayed
Gadolinium-Enhanced MRI Detects Myocardial
Border Zone Injury in a Pig Ischemia-Reperfusion
Model.” Circulation. Cardiovascular Imaging 4 (5):
574–82.
De Groot, D., Grundmann, S., Timmers, L.,
Pasterkamp, G. and Hoefer, I.E. 2011. “Assessment
of Collateral Artery Function and Growth in a Pig
Model of Stepwise Coronary Occlusion.” American
Journal of Physiology. Heart and Circulatory
Physiology 300 (1): H408–14.
Dib, N., Diethrich, E.B., Campbell, A., Gahremanpour,
A., McGarry, M. and Opie, S.R. 2006. “A
Percutaneous Swine Model of Myocardial
Infarction.” Journal of Pharmacological and
Toxicological Methods 53 (3): 256–63.
Díez, J.L., Hernandiz, A., Cosín-Aguilar, J., Aguilar,
A. and Portolés, M. 2013. “Sum of Effects of
Myocardial Ischemia Followed by Electrically
Induced Tachycardia on Myocardial Function.”
Medical Science Monitor Basic Research 19
(January): 153–62.
Dixon, J.A. and Spinale, F.G. 2009. “Large Animal
Models of Heart Failure: A Critical Link in the
Translation of Basic Science to Clinical Practice.”
Circulation. Heart Failure 2 (3): 262–71.
Dixon, J.L., Stoops, J.D., Parker, J.L., Laughlin, M.H.,
Weisman, G.A. and Sturek, M. 1999. “Dyslipidemia
and Vascular Dysfunction in Diabetic Pigs Fed an
Atherogenic Diet.” Arteriosclerosis, Thrombosis,
and Vascular Biology 19 (12): 2981–92.
Doganci, S., Yildirim, V., Bolcal, C., Korkusuz, P.,
Gumusel, B., Demirkilic, U. and Aydin, A. 2012.
“Sodium Nitrite and Cardioprotective Effect in Pig
Regional Myocardial Ischemia-Reperfusion Injury
Model.” Advances in Clinical and Experimental
Medicine : Official Organ Wroclaw Medical
University 21 (6): 713–26.
Dogné, J.-M., Rolin, S., Pétein, M., Tchana-Sato, V.,
Ghuysen, A., Lambermont, B., Hanson, J. et al.
2005. “Characterization of an Original Model of
Myocardial Infarction Provoked by Coronary Artery
Thrombosis Induced by Ferric Chloride in Pig.”
Thrombosis Research 116 (5): 431–42.
Duran, J.M., Taghavi, S., Berretta, R.M., Makarewich,
C.A., Sharp Iii, T., Starosta, T., Udeshi, F.,
George, J.C., Kubo, H. and Houser, S.R. 2012. “A
Characterization and Targeting of the Infarct Border
Zone in a Swine Model of Myocardial Infarction.”
Clinical and Translational Science 5 (5): 416–21.
Dyson, M.C., Alloosh, M., Vuchetich, J.P., Mokelke,
E.A. and Sturek, M. 2006. “Components of
Metabolic Syndrome and Coronary Artery Disease
in Female Ossabaw Swine Fed Excess Atherogenic
Diet.” Comparative Medicine 56 (1): 35–45.
Fallavollita, J.A. 2000. “Spatial Heterogeneity in
Fasting and Insulin-Stimulated 18F-2-Deoxyglucose
Uptake in Pigs With Hibernating Myocardium.”
Circulation 102 (8): 908–14.
Fallavollita, J.A. 2002. “Coronary Patency and Its
Relation to Contractile Reserve in Hibernating
Myocardium.” Cardiovascular Research 55 (1):
131–40.
Fallavollita, J.A. and Canty, J.M. 2002. “Ischemic
Cardiomyopathy in Pigs with Two-Vessel
Occlusion and Viable, Chronically Dysfunctional
Myocardium.” American Journal of Physiology.
References59
Heart and Circulatory Physiology 282 (4): H1370–
79.
Fallavollita, J.A., Jacob, S., Young, R.F. and Canty,
J.M. 1999. “Regional Alterations in SR Ca(2+)ATPase, Phospholamban, and HSP-70 Expression in
Chronic Hibernating Myocardium.” The American
Journal of Physiology 277 (4 Pt 2): H1418–28.
Gálvez-Montón, C., Prat-Vidal, C., Díaz-Güemes, I.,
Crisóstomo, V., Soler-Botija, C., Roura, S., LluciàValldeperas, A., Perea-Gil, I., Sánchez-Margallo,
F.M. and Bayes-Genis, A. 2014. “Comparison of Two
Preclinical Myocardial Infarct Models: Coronary
Coil Deployment versus Surgical Ligation.” Journal
of Translational Medicine 12: 137.
Fallavollita, J.A., Lim, H. and Canty, J.M. 2001.
“Myocyte Apoptosis and Reduced SR Gene
Expression Precede the Transition from Chronically
Stunned to Hibernating Myocardium.” Journal of
Molecular and Cellular Cardiology 33 (11): 1937–
44.
Gálvez-Montón, C., Prat-Vidal, C., Roura, S., SolerBotija, C., Llucià-Valldeperas, A., Díaz-Güemes, I.,
Sánchez-Margallo, F.M. and Bayes-Genis, A. 2013.
“Post-Infarction Scar Coverage Using a PericardialDerived Vascular Adipose Flap. Pre-Clinical
Results.” International Journal of Cardiology 166
(2): 469–74.
Fallavollita, J.A., Logue, M. and Canty, J.M. 2001.
“Stability of Hibernating Myocardium in Pigs with a
Chronic Left Anterior Descending Coronary Artery
Stenosis: Absence of Progressive Fibrosis in the
Setting of Stable Reductions in Flow, Function and
Coronary Flow Reserve.” Journal of the American
College of Cardiology 37 (7): 1989–95.
Garcia-Dorado, D., Theroux, P., Elizaga, J., Galinanes,
M., Solares, J., Riesgo, M., Gomez, M.J., GarciaDorado, A. and Aviles, F.F. 1987. “Myocardial
Reperfusion in the Pig Heart Model: Infarct Size and
Duration of Coronary Occlusion.” Cardiovascular
Research 21 (7): 537–44.
Fallavollita, J.A., Perry, B.J. and Canty, J.M. 1997.
“18F-2-Deoxyglucose Deposition and Regional
Flow in Pigs With Chronically Dysfunctional
Myocardium : Evidence for Transmural Variations
in Chronic Hibernating Myocardium.” Circulation
95 (7): 1900–1909.
Fallavollita, J.A., Riegel, B.J., Suzuki, G., Valeti, U.
and Canty, J.M. 2005. “Mechanism of Sudden
Cardiac Death in Pigs with Viable Chronically
Dysfunctional
Myocardium
and
Ischemic
Cardiomyopathy.” American Journal of Physiology.
Heart and Circulatory Physiology 289 (6): H2688–
96.
Finegold, J.A., Asaria, P. and Francis, D.P. 2013.
“Mortality from Ischaemic Heart Disease by Country,
Region, and Age: Statistics from World Health
Organisation and United Nations.” International
Journal of Cardiology 168 (2): 934–45.
Fish, K.M., Ladage, D., Kawase, Y., Karakikes,
I., Jeong, D., Ly, H., Ishikawa, K. et al. 2013.
“AAV9.I-1c Delivered via Direct Coronary Infusion
in a Porcine Model of Heart Failure Improves
Contractility and Mitigates Adverse Remodeling.”
Circ.Heart Fail. 6: 310–17.
Francis, G.S., Bartos, J.A. and Adatya, S. 2014.
“Inotropes.” Journal of the American College of
Cardiology 63 (20): 2069–78.
Galiuto, L., May-Newman, K., del Balzo, U., Flaim,
S.F., Iliceto, S. and DeMaria, A.N. 2002. “Assessment
of Coronary Stenoses of Graded Severity by
Myocardial Contrast Echocardiography.” Journal of
the American Society of Echocardiography 15 (3):
197–205.
Gelsomino, S., Lucà, F., Nediani, C., Orlandini, S.Z.,
Bani, D., Rubino, A.S., Renzulli, A. et al. 2013.
“Early Hemodynamic and Biochemical Changes in
Overloaded Swine Ventricle.” Texas Heart Institute
Journal / from the Texas Heart Institute of St. Luke’s
Episcopal Hospital, Texas Children’s Hospital 40
(3): 235–45.
Gelsomino, S., Lucà, F., Renzulli, A., Rubino, A.S.,
Romano, S.M., van der Veen, F.H., Carella, R.,
Maessen, J.G., Gensini, G.F. and Lorusso, R. 2011.
“Increased Coronary Blood Flow and Cardiac
Contractile Efficiency with Intraaortic Balloon
Counterpulsation in a Porcine Model of Myocardial
Ischemia-Reperfusion Injury.” ASAIO Journal
(American Society for Artificial Internal Organs :
1992) 57 (5): 375–81.
Gelsomino, S., Renzulli, A., Rubino, A.S., Romano,
S.M., Lucà, F., Valente, S., Gensini, G.F. and
Lorusso, R. 2012. “Effects of 1:1, 1:2 or 1:3 IntraAortic Balloon Counterpulsation/heart Support on
Coronary Haemodynamics and Cardiac Contractile
Efficiency in an Animal Model of Myocardial
Ischaemia/reperfusion.” European Journal of
Cardio-Thoracic Surgery : Official Journal of the
European Association for Cardio-Thoracic Surgery
42 (2): 325–32; discussion 332.
George, R.T., Mehra, V.C., Saraste, A. and Knuuti, J.
2012. “Myocardial Perfusion by CT versus Hybrid
Imaging.” Cardiology Clinics 30 (1): 135–46.
Gerrity, R.G. 1981a. “The Role of the Monocyte
in Atherogenesis: I. Transition of Blood-Borne
Monocytes into Foam Cells in Fatty Lesions.” The
American Journal of Pathology 103 (2): 181–90.
60
References
Gerrity, R.G. 1981b. “The Role of the Monocyte in
Atherogenesis: II. Migration of Foam Cells from
Atherosclerotic Lesions.” The American Journal of
Pathology 103 (2): 191–200.
Gerrity, R.G., Goss, J.A. and Soby, L. 1985. “Control
of Monocyte Recruitment by Chemotactic
Factor(s) in Lesion-Prone Areas of Swine Aorta.”
Arteriosclerosis 5 (1): 55–66.
Gerrity, R.G., Natarajan, R., Nadler, J.L. and
Kimsey, T. 2001. “Diabetes-Induced Accelerated
Atherosclerosis in Swine.” Diabetes 50 (7): 1654–
65.
Gewirtz, H., Brautigan, D.L., Olsson, R.A., Brown,
P. and Most, A.S. 1983. “Role of Adenosine in the
Maintenance of Coronary Vasodilation Distal to a
Severe Coronary Artery Stenosis. Observations in
Conscious Domestic Swine.” Circulation Research
53 (1): 42–51.
Gewirtz, H. and Most, A.S. 1981. “Production of a
Critical Coronary Arterial Stenosis in Closed Chest
Laboratory Animals.” The American Journal of
Cardiology 47 (3): 589–96.
Gewirtz, H., Williams, D.O. and Most, A.S. 1983.
“Quantitative Assessment of the Effects of a
Fixed 50% Coronary Artery Stenosis on Regional
Myocardial Flow Reserve and Transmural
Distribution of Blood Flow.” Journal of the
American College of Cardiology 1 (5): 1273–80.
Giordano, C., Thorn, S.L., Renaud, J.M., Al-Atassi,
T., Boodhwani, M., Klein, R., Kuraitis, D. et al.
2013. “Preclinical Evaluation of BiopolymerDelivered Circulating Angiogenic Cells in a Swine
Model of Hibernating Myocardium.” Circulation.
Cardiovascular Imaging 6 (6): 982–91.
Gould, K.L., Lipscomb, K. and Hamilton, G.W. 1974.
“Physiologic Basis for Assessing Critical Coronary
Stenosis.” The American Journal of Cardiology 33
(1): 87–94.
Granada, J.F., Milewski, K., Zhao, H., Stankus, J.J.,
Tellez, A., Aboodi, M.S., Kaluza, G.L. et al. 2011.
“Vascular Response to Zotarolimus-Coated Balloons
in Injured Superficial Femoral Arteries of the
Familial Hypercholesterolemic Swine.” Circulation.
Cardiovascular Interventions 4 (5): 447–55.
Granada, J.F., Moreno, P.R., Burke, A.P., Schulz, D.G.,
Raizner, A.E. and Kaluza, G.L. 2005. “Endovascular
Needle Injection of Cholesteryl Linoleate into the
Arterial Wall Produces Complex Vascular Lesions
Identifiable by Intravascular Ultrasound: Early
Development in a Porcine Model of Vulnerable
Plaque.” Coronary Artery Disease 16 (4): 217–24.
Greupner, J., Zimmermann, E., Grohmann, A., Dübel,
H.-P., Althoff, T.F., Althoff, T., Borges, A.C. et al.
2012. “Head-to-Head Comparison of Left Ventricular
Function Assessment with 64-Row Computed
Tomography, Biplane Left Cineventriculography,
and Both 2- and 3-Dimensional Transthoracic
Echocardiography: Comparison with Magnetic
Resonance Imaging as the Reference S.” Journal
of the American College of Cardiology 59 (21):
1897–1907.
Grinstead, W.C., Rodgers, G.P., Mazur, W., French,
B.A., Cromeens, D., Van Pelt, C., West, S.M. and
Raizner, A.E. 1994. “Comparison of Three Porcine
Restenosis Models: The Relative Importance of
Hypercholesterolemia, Endothelial Abrasion, and
Stenting.” Coronary Artery Disease 5 (5): 425–34.
Gross, G.J. and Auchampach, J.A. 1992. “Blockade
of ATP-Sensitive Potassium Channels Prevents
Myocardial Preconditioning in Dogs.” Circ.Res. 70:
223–33.
Hagar, J.M., Hale, S.L. and Kloner, R.A. 1991.
“Effect of Preconditioning Ischemia on Reperfusion
Arrhythmias after Coronary Artery Occlusion and
Reperfusion in the Rat.” Circulation Research 68
(1): 61–68.
Hamamdzic, D., Fenning, R.S., Patel, D., Mohler,
E.R., Orlova, K.A., Wright, A.C., Llano, R. et
al. 2010. “Akt Pathway Is Hypoactivated by
Synergistic Actions of Diabetes Mellitus and
Hypercholesterolemia Resulting in Advanced
Coronary Artery Disease.” American Journal of
Physiology. Heart and Circulatory Physiology 299
(3): H699–706.
Hansson, G.K. and Libby, P. 2006. “The Immune
Response in Atherosclerosis: A Double-Edged
Sword.” Nature Reviews. Immunology 6 (7): 508–
19.
Hardt, S.E., Pekrul, I., Hansen, A., Gebhard,
M.M., Kuebler, W. and Kuecherer, H.F. 2001.
“Differential Value of Adenosine Myocardial
Contrast Echocardiography and Dobutamine
Stress Echocardiography in Evaluating Functional
Significance of Coronary Artery Stenosis in a
Porcine Model.” Basic Research in Cardiology 96
(4): 415–21.
Hashizume, R., Fujimoto, K.L., Hong, Y., Guan,
J., Toma, C., Tobita, K. and Wagner, W.R. 2013.
“Biodegradable Elastic Patch Plasty Ameliorates
Left Ventricular Adverse Remodeling after IschemiaReperfusion Injury: A Preclinical Study of a Porous
Polyurethane Material in a Porcine Model.” The
Journal of Thoracic and Cardiovascular Surgery
146 (2): 391–99.e1.
Hasler-Rapacz, J., Ellegren, H., Fridolfsson, A.K.,
Kirkpatrick, B., Kirk, S., Andersson, L. and Rapacz,
J. 1998. “Identification of a Mutation in the Low
References61
Density Lipoprotein Receptor Gene Associated
with Recessive Familial Hypercholesterolemia in
Swine.” American Journal of Medical Genetics 76
(5): 379–86.
Hasler-Rapacz, J., Prescott, M.F., Von Linden-Reed,
J., Rapacz, J.M., Hu, Z. and Rapacz, J. 1995.
“Elevated Concentrations of Plasma Lipids and
Apolipoproteins B, C-III, and E Are Associated
with the Progression of Coronary Artery Disease
in
Familial
Hypercholesterolemic
Swine.”
Arteriosclerosis, Thrombosis, and Vascular Biology
15 (5): 583–92.
Heilmann, C., Kostic, C., Giannone, B., Grawitz,
A.B., Armbruster, W., Lutter, G., Beyersdorf,
F. and Göbel, H. 2006. “Improvement of
Contractility Accompanies Angiogenesis rather than
Arteriogenesis in Chronic Myocardial Ischemia.”
Vascular Pharmacology 44 (5): 326–32.
Heinonen, S.E., Leppänen, P., Kholová, I., Lumivuori,
H., Häkkinen, S.-K., Bosch, F., Laakso, M. and
Ylä-Herttuala, S. 2007. “Increased Atherosclerotic
Lesion Calcification in a Novel Mouse Model
Combining Insulin Resistance, Hyperglycemia, and
Hypercholesterolemia.” Circulation Research 101
(10): 1058–67.
Hemetsberger, R., Farhan, S., Strehblow, C., Sperker,
W., Pavo, I., Petrasi, Z., Hemetsberger, H. et al.
2008. “Association between the Efficacy of Dual
Antiplatelet Therapy and the Development of inStent Neointimal Hyperplasia in Porcine Coronary
Arteries.” Coronary Artery Disease 19 (8): 635–43.
Heusch, G., Libby, P., Gersh, B., Yellon, D., Böhm, M.,
Lopaschuk, G. and Opie, L. 2014. “Cardiovascular
Remodelling in Coronary Artery Disease and Heart
Failure.” Lancet 383 (9932): 1933–43.
Hinkel, R., Penzkofer, D., Zühlke, S., Fischer, A.,
Husada, W., Xu, Q.-F., Baloch, E. et al. 2013.
“Inhibition of microRNA-92a Protects against
Ischemia/reperfusion Injury in a Large-Animal
Model.” Circulation 128 (10): 1066–75.
Holz, A., Lautamäki, R., Sasano, T., Merrill, J.,
Nekolla, S.G., Lardo, A.C. and Bengel, F.M. 2009.
“Expanding the Versatility of Cardiac PET/CT:
Feasibility of Delayed Contrast Enhancement CT
for Infarct Detection in a Porcine Model.” Journal
of Nuclear Medicine : Official Publication, Society
of Nuclear Medicine 50 (2): 259–65.
Hong, X., Fan, H., Lu, R., Chan, P. and Liu, Z. 2013.
“Merit of Anisodamine Combined with Opioid
Δ-Receptor Activation in the Protection against
Myocardial Injury during Cardiopulmonary
Bypass.” BioMed Research International 2013
(January): 212801.
Horstick, G., Bierbach, B., Abegunewardene, N.,
Both, S., Kuhn, S., Manefeld, D., Reinecke, H.-J.
et al. 2009. “Critical Single Proximal Left Arterial
Descending Coronary Artery Stenosis to Mimic
Chronic Myocardial Ischemia: A New Model
Induced by Minimal Invasive Technology.” Journal
of Vascular Research 46 (4): 290–98.
Hsiao, Y.-M., Mathias, C.J., Wey, S.-P., Fanwick,
P.E. and Green, M.A. 2009. “Synthesis and
Biodistribution of Lipophilic and Monocationic
Gallium
Radiopharmaceuticals
Derived
from
N,N’-bis(3-Aminopropyl)-N,N’Dimethylethylenediamine: Potential Agents for PET
Myocardial Imaging with 68Ga.” Nuclear Medicine
and Biology 36 (1): 39–45.
Huang, Z., Ge, J., Sun, A., Wang, Y., Zhang, S., Cui,
J., Qian, J. and Zou, Y. 2010. “Ligating LAD with
Its Whole Length rather than Diagonal Branches
as Coordinates Is More Advisable in Establishing
Stable Myocardial Infarction Model of Swine.” Exp.
Anim 59: 431–39.
Hughes, G.C., Landolfo, C.K., Yin, B., DeGrado,
T.R., Coleman, R.E., Landolfo, K.P. and Lowe,
J.E. 2001. “Is Chronically Dysfunctional yet Viable
Myocardium Distal to a Severe Coronary Stenosis
Hypoperfused?” The Annals of Thoracic Surgery 72
(1): 163–68.
Hughes, G.C., Post, M.J., Simons, M. and Annex, B.H.
2003. “Translational Physiology: Porcine Models
of Human Coronary Artery Disease: Implications
for Preclinical Trials of Therapeutic Angiogenesis.”
Journal of Applied Physiology (Bethesda, Md. :
1985) 94 (5): 1689–1701.
Iida, H., Rhodes, C.G., De, S.R., Araujo, L.I.,
Bloomfield, P.M., Lammertsma, A.A. and Jones, T.
1992. “Use of the Left Ventricular Time-Activity
Curve as a Noninvasive Input Function in Dynamic
Oxygen-15-Water Positron Emission Tomography.”
J.Nucl.Med. 33: 1669–77.
Iida, H., Ruotsalainen, U., Mäki, M., Haaparnata,
M., Bergman, J., Voipio-Pulkki, L.-M., Nuutila,
P., Koshino, K. and Knuuti, J. 2012. “F-18
Fluorodeoxyglucose Uptake and Water-Perfusable
Tissue Fraction in Assessment of Myocardial
Viability.” Annals of Nuclear Medicine 26 (8): 644–
55.
Ikonen, T.S., Pätilä, T., Virtanen, K., Lommi, J.,
Lappalainen, K., Kankuri, E., Krogerus, L. and
Harjula, A. 2007. “Ligation of Ameroid-Stenosed
Coronary Artery Leads to Reproducible Myocardial
Infarction--a Pilot Study in a Porcine Model.” The
Journal of Surgical Research 142 (1): 195–201.
Ishikawa, K., Ladage, D., Takewa, Y., Yaniz, E.,
Chen, J., Tilemann, L., Sakata, S., Badimon, J.J.,
62
References
Hajjar, R.J. and Kawase, Y. 2011. “Development of
a Preclinical Model of Ischemic Cardiomyopathy
in Swine.” Am.J.Physiol Heart Circ.Physiol 301:
H530–37.
“An Experimental Method for the Percutaneous
Induction of a Posterolateral Infarct and Functional
Ischemic Mitral Regurgitation.” The Journal of
Heart Valve Disease 14 (4): 460–66.
Jager, N.A., Westra, J., Golestani, R., van Dam, G.M.,
Low, P.S., Tio, R.A., Slart, R.H.J.A., Boersma,
H.H., Bijl, M. and Zeebregts, C.J. 2014. “Folate
Receptor-Β Imaging Using 99mTc-Folate to Explore
Distribution of Polarized Macrophage Populations
in Human Atherosclerotic Plaque.” Journal of
Nuclear Medicine : Official Publication, Society of
Nuclear Medicine 55 (12): 1945–51.
Jugdutt, B.I., Hutchins, G.M., Bulkley, B.H. and
Becker, L.C. 1979. “Myocardial Infarction in the
Conscious Dog: Three-Dimensional Mapping
of Infarct, Collateral Flow and Region at Risk.”
Circulation 60 (5): 1141–50.
Jiang, Y., Chang, P., Pei, Y., Li, B., Liu, Y., Zhang, Z.,
Yu, J., Zhu, D. and Liu, X. 2014. “Intramyocardial
Injection of Hypoxia-Preconditioned AdiposeDerived Stromal Cells Treats Acute Myocardial
Infarction: An in Vivo Study in Swine.” Cell and
Tissue Research, August.
Johanna Haukkala, Iina Laitinen, Pauliina Luoto, Peter
Iveson, Ian Wilson, Hege Karlsen, Alan Cuthbertson
et al. 2009. “68Ga-DOTA-RGD Peptide:
Biodistribution and Binding into Atherosclerotic
Plaques in Mice.” European Journal of Nuclear
Medicine and Molecular Imaging 36 (12): 2058–67.
Johnson, L.L., Schofield, L., Mastrofrancesco, P.,
Donahay, T. and Nott, L. 1998. “Technetium99m-Nitroimadazole Uptake in a Swine Model of
Demand Ischemia.” Journal of Nuclear Medicine :
Official Publication, Society of Nuclear Medicine 39
(8): 1468–75.
Johnson, L.L., Schofield, L.M., Verdesca, S.A., Sharaf,
B.L., Jones, R.M., Virmani, R. and Khaw, B.A.
2000. “In Vivo Uptake of Radiolabeled Antibody
to Proliferating Smooth Muscle Cells in a Swine
Model of Coronary Stent Restenosis.” Journal of
Nuclear Medicine : Official Publication, Society of
Nuclear Medicine 41 (9): 1535–40.
Jormalainen, M., Vento, A.E., Lukkarinen, H., Kääpä,
P., Kytö, V., Lauronen, J., Paavonen, T., SuojarantaYlinen, R. and Petäjä, J. 2007. “Inhibition of
Thrombin during Reperfusion Improves Immediate
Postischemic Myocardial Function and Modulates
Apoptosis in a Porcine Model of Cardiopulmonary
Bypass.” Journal of Cardiothoracic and Vascular
Anesthesia 21 (2): 224–31.
Joshi, N. V, Vesey, A.T., Williams, M.C., Shah, A.S. V,
Calvert, P.A., Craighead, F.H.M., Yeoh, S.E. et al.
2014. “18F-Fluoride Positron Emission Tomography
for Identification of Ruptured and High-Risk
Coronary Atherosclerotic Plaques: A Prospective
Clinical Trial.” Lancet 383 (9918): 705–13.
Joudinaud, T.M., Kegel, C.L., Gabster, A.A., Sanz,
M.L., MacDonald, A., Propp, D., Callaghan, E.,
Weber, P.A., Hvass, U. and Duran, C.M.G. 2005.
Kamimura, R., Suzuki, S., Nozaki, S., Sakamoto, H.,
Maruno, H. and Kawaida, H. 1996. “Branching
Patterns in Coronary Artery and Ischemic Areas
Induced by Coronary Arterial Occlusion in the
CLAWN Miniature Pig.” Exp.Anim 45 (1341-1357
(Print)). Institute of Laboratory Animal Sciences,
Faculty of Medicine, Kagoshima University, Japan:
149–53.
Kanlop, N., Thommasorn, S., Palee, S.,
Weerateerangkul, P., Suwansirikul, S., Chattipakorn,
S. and Chattipakorn, N. 2011. “Granulocyte
Colony-Stimulating Factor Stabilizes Cardiac
Electrophysiology and Decreases Infarct Size during
Cardiac Ischaemic/reperfusion in Swine.” Acta
Physiologica (Oxford, England) 202 (1): 11–20.
Kassab, G.S., Berkley, J. and Fung, Y.C. 1997.
“Analysis of Pig’s Coronary Arterial Blood
Flow with Detailed Anatomical Data.” Annals of
Biomedical Engineering 25 (1): 204–17.
Kawamura, M., Miyagawa, S., Miki, K., Saito, A.,
Fukushima, S., Higuchi, T., Kawamura, T. et al.
2012. “Feasibility, Safety, and Therapeutic Efficacy
of Human Induced Pluripotent Stem Cell-Derived
Cardiomyocyte Sheets in a Porcine Ischemic
Cardiomyopathy Model.” Circulation 126 (11 Suppl
1): S29–37.
Kelly, J.D., Forster, A.M., Higley, B., Archer, C.M.,
Booker, F.S., Canning, L.R., Chiu, K.W., Edwards,
B., Gill, H.K. and McPartlin, M. 1993. “Technetium99m-Tetrofosmin as a New Radiopharmaceutical for
Myocardial Perfusion Imaging.” Journal of Nuclear
Medicine : Official Publication, Society of Nuclear
Medicine 34 (2): 222–27.
Kersten, J.R., Montgomery, M.W., Pagel, P.S. and
Warltier, D.C. 2000. “Levosimendan, a New Positive
Inotropic Drug, Decreases Myocardial Infarct Size
via Activation of K(ATP) Channels.” Anesth.Analg.
90: 5–11.
Kim, D.N., Schmee, J., Baker, J.E., Lunden, G.M.,
Sheehan, C.E., Lee, C.S., Eastman, A., Solis, O.,
Ross, J.S. and Thomas, W.A. 1993. “Dietary Fish Oil
Reduces Microthrombi over Atherosclerotic Lesions
in Hyperlipidemic Swine Even in the Absence of
Plasma Cholesterol Reduction.” Experimental and
Molecular Pathology 59 (2): 122–35.
References63
Kim, R.J., Wu, E., Rafael, A., Chen, E.L., Parker,
M.A., Simonetti, O., Klocke, F.J., Bonow, R.O. and
Judd, R.M. 2000. “The Use of Contrast-Enhanced
Magnetic Resonance Imaging to Identify Reversible
Myocardial Dysfunction.” The New England
Journal of Medicine 343 (20): 1445–53.
Kraitchman, D.L., Chin, B.B., Heldman, A.W.,
Solaiyappan, M. and Bluemke, D.A. 2002. “MRI
Detection of Myocardial Perfusion Defects due to
Coronary Artery Stenosis with MS-325.” Journal
of Magnetic Resonance Imaging : JMRI 15 (2):
149–58.
Kim, S.J., Kudej, R.K., Yatani, A., Kim, Y.K., Takagi,
G., Honda, R., Colantonio, D.A. et al. 2001.
“A Novel Mechanism for Myocardial Stunning
Involving Impaired Ca(2+) Handling.” Circulation
Research 89 (9): 831–37.
Kreutz, R.P., Alloosh, M., Mansour, K., Neeb, Z.,
Kreutz, Y., Flockhart, D.A. and Sturek, M. 2011.
“Morbid Obesity and Metabolic Syndrome in
Ossabaw Miniature Swine Are Associated with
Increased Platelet Reactivity.” Diabetes, Metabolic
Syndrome and Obesity : Targets and Therapy 4:
99–105.
Kim, W., Jeong, M.H., Sim, D.S., Hong, Y.J., Song,
H.C., Park, J.T. and Ahn, Y.K. 2011. “A Porcine
Model of Ischemic Heart Failure Produced by
Intracoronary Injection of Ethyl Alcohol.” Heart
and Vessels 26 (3): 342–48.
Kiugel, M., Dijkgraaf, I., Kytö, V., Helin, S., Liljenbäck,
H., Saanijoki, T., Yim, C.-B. et al. 2014. “Dimeric
[(68)Ga]DOTA-RGD Peptide Targeting Αvβ 3
Integrin Reveals Extracellular Matrix Alterations
after Myocardial Infarction.” Molecular Imaging
and Biology : MIB : The Official Publication of the
Academy of Molecular Imaging, July.
Knaapen, P., Germans, T., Knuuti, J., Paulus, W.J.,
Dijkmans, P.A., Allaart, C.P., Lammertsma, A.A.
and Visser, F.C. 2007. “Myocardial Energetics
and Efficiency: Current Status of the Noninvasive
Approach.” Circulation 115: 918–27.
Knuuti, J., Kajander, S., Mäki, M. and Ukkonen, H.
2009. “Quantification of Myocardial Blood Flow
Will Reform the Detection of CAD.” Journal of
Nuclear Cardiology : Official Publication of the
American Society of Nuclear Cardiology 16 (4):
497–506.
Knuuti, J., Schelbert, H.R. and Bax, J.J. 2002. “The
Need for Standardisation of Cardiac FDG PET
Imaging in the Evaluation of Myocardial Viability
in Patients with Chronic Ischaemic Left Ventricular
Dysfunction.” European Journal of Nuclear
Medicine and Molecular Imaging 29 (9): 1257–66.
Koudstaal, S., Jansen Of Lorkeers, S.J., van Slochteren,
F.J., van der Spoel, T.I.G., van de Hoef, T.P., Sluijter,
J.P., Siebes, M., Doevendans, P.A., Piek, J.J. and
Chamuleau, S.A.J. 2013. “Assessment of Coronary
Microvascular Resistance in the Chronic Infarcted
Pig Heart.” Journal of Cellular and Molecular
Medicine 17 (9): 1128–35.
Kraitchman, D.L., Bluemke, D.A., Chin, B.B. and
Heldman, A.W. 2000. “A Minimally Invasive
Method for Creating Coronary Stenosis in a Swine
Model for MRI and SPECT Imaging.” Investigative
Radiology 35 (7): 445–51.
Krombach, G.A., Kinzel, S., Mahnken, A.H., Günther,
R.W. and Buecker, A. 2005. “Minimally Invasive
Close-Chest Method for Creating Reperfused
or Occlusive Myocardial Infarction in Swine.”
Investigative Radiology 40 (1): 14–18.
Kuster, D.W.D., Merkus, D., Kremer, A., van Ijcken,
W.F.J., de Beer, V.J., Verhoeven, A.J.M. and
Duncker, D.J. 2011. “Left Ventricular Remodeling in
Swine after Myocardial Infarction: A Transcriptional
Genomics Approach.” Basic Research in Cardiology
106 (6): 1269–81.
Kuzuya, T., Hoshida, S., Yamashita, N., Fuji, H.,
Oe, H., Hori, M., Kamada, T. and Tada, M. 1993.
“Delayed Effects of Sublethal Ischemia on the
Acquisition of Tolerance to Ischemia.” Circulation
Research 72 (6): 1293–99.
Lai, T., Fallon, J.T., Liu, J., Mangion, J., Gillam,
L., Waters, D. and Chen, C. 2000. “Reversibility
and Pathohistological Basis of Left Ventricular
Remodeling
in
Hibernating
Myocardium.”
Cardiovascular Pathology 9 (6): 323–35.
Laitinen, I., Luoto, P., Någren, K., Marjamäki, P.M.,
Silvola, J.M.U., Hellberg, S., Laine, V.J.O., YläHerttuala, S., Knuuti, J. and Roivainen, A. 2010.
“Uptake of 11C-Choline in Mouse Atherosclerotic
Plaques.” Journal of Nuclear Medicine : Official
Publication, Society of Nuclear Medicine 51 (5):
798–802.
Laitinen, I., Marjamäki, P., Någren, K., Laine,
V.J.O., Wilson, I., Leppänen, P., Ylä-Herttuala,
S., Roivainen, A. and Knuuti, J. 2009. “Uptake of
Inflammatory Cell Marker [11C]PK11195 into
Mouse Atherosclerotic Plaques.” European Journal
of Nuclear Medicine and Molecular Imaging 36 (1):
73–80.
Laitinen, I., Saraste, A., Weidl, E., Poethko, T., Weber,
A.W., Nekolla, S.G., Leppänen, P. et al. 2009.
“Evaluation of alphavbeta3 Integrin-Targeted
Positron Emission Tomography Tracer 18F-GalactoRGD for Imaging of Vascular Inflammation in
64
References
Atherosclerotic Mice.” Circulation. Cardiovascular
Imaging 2 (4): 331–38.
Lassaletta, A.D., Chu, L.M. and Sellke, F.W. 2012.
“Effects of Alcohol on Pericardial Adhesion
Formation in Hypercholesterolemic Swine.” The
Journal of Thoracic and Cardiovascular Surgery
143 (4): 953–59.
Laufer, E.M., Reutelingsperger, C.P.M., Narula, J.
and Hofstra, L. 2008. “Annexin A5: An Imaging
Biomarker of Cardiovascular Risk.” Basic Research
in Cardiology 103 (2): 95–104.
Lautamäki, R., Schuleri, K.H., Sasano, T., Javadi,
M.S., Youssef, A., Merrill, J., Nekolla, S.G.,
Abraham, M.R., Lardo, A.C. and Bengel, F.M. 2009.
“Integration of Infarct Size, Tissue Perfusion, and
Metabolism by Hybrid Cardiac Positron Emission
Tomography/computed Tomography: Evaluation
in a Porcine Model of Myocardial Infarction.”
Circulation. Cardiovascular Imaging 2 (4): 299–
305.
Li, X., Shao, D., Wang, G., Jiang, T., Wu, H., Gu,
B., Cao, K., Zhang, J., Qi, L. and Chen, Y. 2014.
“Effects of Different LAD-Blocked Sites on the
Development of Acute Myocardial Infarction and
Malignant Arrhythmia in a Swine Model.” Journal
of Thoracic Disease 6 (9): 1271–77.
Li, X., Yang, Y., Cheng, Y., Dou, K., Tian, Y. and
Meng, X. 2013. “Protein Kinase A-Mediated
Cardioprotection of Tongxinluo Relates to the
Inhibition of Myocardial Inflammation, Apoptosis,
and Edema in Reperfused Swine Hearts.” Chinese
Medical Journal 126 (8): 1469–79.
Liao, R., Nascimben, L., Friedrich, J., Gwathmey, J.K.
and Ingwall, J.S. 1996. “Decreased Energy Reserve
in an Animal Model of Dilated Cardiomyopathy.
Relationship
to
Contractile
Performance.”
Circulation Research 78 (5): 893–902.
Liao, S.-Y., Siu, C.-W., Liu, Y., Zhang, Y., Chan, W.-S.,
Wu, E.X., Wu, Y. et al. 2010. “Attenuation of Left
Ventricular Adverse Remodeling with Epicardial
Patching after Myocardial Infarction.” Journal of
Cardiac Failure 16 (7): 590–98.
Libby, P. 2002. “Inflammation in Atherosclerosis.”
Nature 420 (6917): 868–74.
Libby, P. 2012. “Inflammation in Atherosclerosis.”
Arteriosclerosis, Thrombosis, and Vascular Biology
32 (9): 2045–51.
Libby, P. and Theroux, P. 2005. “Pathophysiology of
Coronary Artery Disease.” Circulation 111 (25):
3481–88.
Liedtke, A.J., Renstrom, B., Nellis, S.H., Hall, J.L.
and Stanley, W.C. 1995. “Mechanical and Metabolic
Functions in Pig Hearts after 4 Days of Chronic
Coronary Stenosis.” Journal of the American
College of Cardiology 26 (3): 815–25.
Liedtke, A.J., Renstrom, B., Nellis, S.H. and
Subramanian, R. 1994. “Myocardial Function and
Metabolism in Pig Hearts after Relief from Chronic
Partial Coronary Stenosis.” Am J Physiol Heart Circ
Physiol 267 (4): H1312–19.
Lim, H., Fallavollita, J.A., Hard, R., Kerr, C.W. and
Canty, J.M. 1999. “Profound Apoptosis-Mediated
Regional Myocyte Loss and Compensatory
Hypertrophy in Pigs with Hibernating Myocardium.”
Circulation 100 (23): 2380–86.
Lim, K.H.H., Halestrap, A.P., Angelini, G.D. and
Suleiman, M.-S. 2005. “Propofol Is Cardioprotective
in a Clinically Relevant Model of Normothermic
Blood Cardioplegic Arrest and Cardiopulmonary
Bypass.” Experimental Biology and Medicine 230
(6): 413–20.
Lin, X., Lee, D. and Wu, D. 2013. “Protective
Effects of NHE1 Inhibition with Sabiporide in an
Experimental Model of Asphyxia-Induced Cardiac
Arrest in Piglets.” Resuscitation 84 (4): 520–25.
Lu, F., Zhao, X., Wu, J., Cui, Y., Mao, Y., Chen, K.,
Yuan, Y., Gong, D., Xu, Z. and Huang, S. 2013.
“MSCs Transfected with Hepatocyte Growth Factor
or Vascular Endothelial Growth Factor Improve
Cardiac Function in the Infarcted Porcine Heart by
Increasing Angiogenesis and Reducing Fibrosis.”
International Journal of Cardiology 167 (6): 2524–
32.
Lukács, E., Magyari, B., Tóth, L., Petrási, Z., Repa, I.,
Koller, A. and Horváth, I. 2012. “Overview of Large
Animal Myocardial Infarction Models (review).”
Acta Physiologica Hungarica 99 (4): 365–81.
Maecke, H.R. and André, J.P. 2007. “68Ga-PET
Radiopharmacy: A Generator-Based Alternative
to 18F-Radiopharmacy.” Ernst Schering Research
Foundation Workshop, no. 62 (January): 215–42.
Mahnken, A.H., Klotz, E., Pietsch, H., Schmidt, B.,
Allmendinger, T., Haberland, U., Kalender, W.A.
and Flohr, T. 2010. “Quantitative Whole Heart Stress
Perfusion CT Imaging as Noninvasive Assessment
of Hemodynamics in Coronary Artery Stenosis:
Preliminary Animal Experience.” Investigative
Radiology 45 (6): 298–305.
Malmberg, M., Vähäsilta, T., Saraste, A., Kytö, V., Kiss,
J., Kentala, E., Kallajoki, M. and Savunen, T. 2006.
“Cardiomyocyte Apoptosis and Duration of Aortic
Clamping in Pig Model of Open Heart Surgery.”
European Journal of Cardio-Thoracic Surgery :
Official Journal of the European Association for
Cardio-Thoracic Surgery 30 (3): 480–84.
References65
Mateo, J., Izquierdo-Garcia, D., Badimon, J.J., Fayad,
Z.A. and Fuster, V. 2014. “Noninvasive Assessment
of Hypoxia in Rabbit Advanced Atherosclerosis
Using 18F-Fluoromisonidazole Positron Emission
Tomographic Imaging.” Circulation. Cardiovascular
Imaging 7 (2): 312–20.
Matter, C.M., Wyss, M.T., Meier, P., Späth, N., von
Lukowicz, T., Lohmann, C., Weber, B. et al. 2006.
“18F-Choline Images Murine Atherosclerotic
Plaques Ex Vivo.” Arteriosclerosis, Thrombosis,
and Vascular Biology 26 (3): 584–89.
Matyal, R., Chu, L., Mahmood, F., Robich, M.P.,
Wang, A., Hess, P.E., Shahul, S., Pinto, D.S.,
Khabbaz, K. and Sellke, F.W. 2012. “Neuropeptide
Y Improves Myocardial Perfusion and Function in a
Swine Model of Hypercholesterolemia and Chronic
Myocardial Ischemia.” Journal of Molecular and
Cellular Cardiology 53 (6): 891–98.
Matyal, R., Sakamuri, S., Wang, A., Mahmood, E.,
Robich, M.P., Khabbaz, K., Hess, P.E., Sellke,
F.W. and Mahmood, F. 2013. “Local Infiltration of
Neuropeptide Y as a Potential Therapeutic Agent
against Apoptosis and Fibrosis in a Swine Model
of Hypercholesterolemia and Chronic Myocardial
Ischemia.” European Journal of Pharmacology 718
(1-3): 261–70.
McDonald, T.O., Gerrity, R.G., Jen, C., Chen, H.-J.,
Wark, K., Wight, T.N., Chait, A. and O’Brien, K.D.
2007. “Diabetes and Arterial Extracellular Matrix
Changes in a Porcine Model of Atherosclerosis.”
The Journal of Histochemistry and Cytochemistry :
Official Journal of the Histochemistry Society 55
(11): 1149–57.
McFalls, E.O., Baldwin, D., Palmer, B., Marx, D.,
Jaimes, D. and Ward, H.B. 1997. “Regional Glucose
Uptake within Hypoperfused Swine Myocardium
as Measured by Positron Emission Tomography.”
The American Journal of Physiology 272 (1 Pt 2):
H343–49.
McFalls, E.O., Sluiter, W., Schoonderwoerd, K.,
Manintveld, O.C., Lamers, J.M.J., Bezstarosti, K.,
van Beusekom, H.M. et al. 2006. “Mitochondrial
Adaptations within Chronically Ischemic Swine
Myocardium.” Journal of Molecular and Cellular
Cardiology 41 (6): 980–88.
McMurray, J.J. V and Pfeffer, M.A. 2005. “Heart
Failure.” Lancet 365 (9474): 1877–89.
Metzsch, C., Liao, Q., Steen, S. and Algotsson, L.
2006. “Myocardial Glycerol Release, Arrhythmias
and Hemodynamic Instability during Regional
Ischemia-Reperfusion in an Open Chest Pig Model.”
Acta Anaesthesiologica Scandinavica 50 (1): 99–
107.
Meyer, M., Bell, S.P., Chen, Z., Nyotowidjojo, I.,
Lachapelle, R.R., Christian, T.F., Gibson, P.C.,
Keating, F.F., Dauerman, H.L. and LeWinter, M.M.
2013. “High Dose Intracoronary N-Acetylcysteine
in a Porcine Model of ST-Elevation Myocardial
Infarction.” Journal of Thrombosis and Thrombolysis
36 (4): 433–41.
Mihaylov, D., van Luyn, M.J. and Rakhorst, G. 2000.
“Development of an Animal Model of Selective
Coronary Atherosclerosis.” Coronary Artery
Disease 11 (2): 145–49.
Mills, I., Fallon, J.T., Wrenn, D., Sasken, H., Gray,
W., Bier, J., Levine, D., Berman, S., Gilson, M. and
Gewirtz, H. 1994. “Adaptive Responses of Coronary
Circulation and Myocardium to Chronic Reduction
in Perfusion Pressure and Flow.” The American
Journal of Physiology 266 (2 Pt 2): H447–57.
Mohler, E.R., Sarov-Blat, L., Shi, Y., Hamamdzic,
D., Zalewski, A., Macphee, C., Llano, R. et
al. 2008. “Site-Specific Atherogenic Gene
Expression Correlates with Subsequent Variable
Lesion Development in Coronary and Peripheral
Vasculature.” Arteriosclerosis, Thrombosis, and
Vascular Biology 28 (5): 850–55.
Munz, M.R., Faria, M.A., Monteiro, J.R., Aguas,
A.P. and Amorim, M.J. 2011. “Surgical Porcine
Myocardial Infarction Model through Permanent
Coronary Occlusion.” Comparative Medicine 61
(5): 445–52.
Murry, C.E., Jennings, R.B. and Reimer, K.A. 1986.
“Preconditioning with Ischemia: A Delay of Lethal
Cell Injury in Ischemic Myocardium.” Circulation
74 (5): 1124–36.
Nahrendorf, M., Keliher, E., Panizzi, P., Zhang, H.,
Hembrador, S., Figueiredo, J.-L., Aikawa, E., Kelly,
K., Libby, P. and Weissleder, R. 2009. “18F-4V
for PET-CT Imaging of VCAM-1 Expression in
Atherosclerosis.” JACC. Cardiovascular Imaging 2
(10): 1213–22.
Nakamura, I., Hasegawa, K., Wada, Y., Hirase, T.,
Node, K. and Watanabe, Y. 2013. “Detection of Early
Stage Atherosclerotic Plaques Using PET and CT
Fusion Imaging Targeting P-Selectin in Low Density
Lipoprotein Receptor-Deficient Mice.” Biochemical
and Biophysical Research Communications 433 (1):
47–51.
Napoli, C., D’Armiento, F.P., Mancini, F.P., Postiglione,
A., Witztum, J.L., Palumbo, G. and Palinski, W.
1997. “Fatty Streak Formation Occurs in Human
Fetal Aortas and Is Greatly Enhanced by Maternal
Hypercholesterolemia. Intimal Accumulation of
Low Density Lipoprotein and Its Oxidation Precede
Monocyte Recruitment into Early Atherosclerotic
66
References
Lesions.” The Journal of Clinical Investigation 100
(11): 2680–90.
PET for in Vivo Atherosclerosis Imaging.” Annals
of Nuclear Medicine 27 (9): 847–54.
Neeb, Z.P., Edwards, J.M., Alloosh, M., Long, X.,
Mokelke, E.A. and Sturek, M. 2010. “Metabolic
Syndrome and Coronary Artery Disease in Ossabaw
Compared with Yucatan Swine.” Comparative
Medicine 60 (4): 300–315.
Papp, Z., Édes, I., Fruhwald, S., De Hert, S.G.,
Salmenperä, M., Leppikangas, H., Mebazaa, A. et
al. 2012. “Levosimendan: Molecular Mechanisms
and Clinical Implications: Consensus of Experts
on the Mechanisms of Action of Levosimendan.”
International Journal of Cardiology 159 (2): 82–87.
Näslund, U., Häggmark, S., Johansson, G., Marklund,
S.L. and Reiz, S. 1992. “A Closed-Chest Myocardial
Occlusion-Reperfusion Model in the Pig:
Techniques, Morbidity and Mortality.” European
Heart Journal 13 (9): 1282–89.
Näslund, U., Häggmark, S., Johansson, G., Pennert,
K., Reiz, S. and Marklund, S.L. 1992. “Effects
of Reperfusion and Superoxide Dismutase on
Myocardial Infarct Size in a Closed Chest Pig
Model.” Cardiovascular Research 26 (2): 170–78.
Ogura, Y., Ouchi, N., Ohashi, K., Shibata, R., Kataoka,
Y., Kambara, T., Kito, T. et al. 2012. “Therapeutic
Impact of Follistatin-like 1 on Myocardial Ischemic
Injury in Preclinical Models.” Circulation 126 (14):
1728–38.
Ohtsuka, S., Ishikawa, K., Suzuki, S., Yamaguchi,
I., Masuda, N., Wada, K. and Uchid, W. 2003. “A
Porcine Model of Ischemic Heart Failure Produced
by Chronic Placement of a Tube in a Coronary
Artery.” European Journal of Heart Failure 5 (5):
591–98.
Olson, A.K., Bouchard, B., Ning, X.-H., Isern,
N., Rosiers, C. Des and Portman, M.A. 2012.
“Triiodothyronine Increases Myocardial Function
and Pyruvate Entry into the Citric Acid Cycle after
Reperfusion in a Model of Infant Cardiopulmonary
Bypass.” American Journal of Physiology. Heart
and Circulatory Physiology 302 (5): H1086–93.
Osipov, R.M., Robich, M.P., Feng, J., Liu, Y.,
Clements, R.T., Glazer, H.P., Sodha, N.R., Szabo,
C., Bianchi, C. and Sellke, F.W. 2009. “Effect of
Hydrogen Sulfide in a Porcine Model of Myocardial
Ischemia-Reperfusion: Comparison of Different
Administration Regimens and Characterization of
the Cellular Mechanisms of Protection.” Journal of
Cardiovascular Pharmacology 54 (4): 287–97.
Oyamada, S., Bianchi, C., Takai, S., Robich, M.P.,
Clements, R.T., Chu, L. and Sellke, F.W. 2010.
“Impact of Acute Myocardial Ischemia Reperfusion
on the Tissue and Blood-Borne Renin-Angiotensin
System.” Basic Research in Cardiology 105 (4):
513–22.
Paeng, J.C., Lee, Y.-S., Lee, J.S., Jeong, J.M., Kim,
K.-B., Chung, J.-K. and Lee, D.S. 2013. “Feasibility
and Kinetic Characteristics of (68)Ga-NOTA-RGD
Pavo, N., Zimmermann, M., Pils, D., Mildner, M.,
Petrási, Z., Petneházy, Ö., Fuzik, J. et al. 2014.
“Long-Acting Beneficial Effect of Percutaneously
Intramyocardially Delivered Secretome of Apoptotic
Peripheral Blood Cells on Porcine Chronic Ischemic
Left Ventricular Dysfunction.” Biomaterials 35 (11):
3541–50.
Pennell, D.J., Sechtem, U.P., Higgins, C.B., Manning,
W.J., Pohost, G.M., Rademakers, F.E., van Rossum,
A.C., Shaw, L.J. and Yucel, E.K. 2004. “Clinical
Indications for Cardiovascular Magnetic Resonance
(CMR): Consensus Panel Report.” European Heart
Journal 25 (21): 1940–65.
Pérez de Prado, A., Cuellas-Ramón, C., RegueiroPurriños, M., Gonzalo-Orden, J.M., Pérez-Martínez,
C., Altónaga, J.R., García-Iglesias, M.J., OrdenRecio, M.A., García-Marín, J.F. and FernándezVázquez, F. 2009. “Closed-Chest Experimental
Porcine Model of Acute Myocardial InfarctionReperfusion.” Journal of Pharmacological and
Toxicological Methods 60 (3): 301–6.
Peukert, D., Laule, M., Kaufels, N., Schnorr, J.,
Taupitz, M., Hamm, B. and Dewey, M. 2009.
“A Minimally Invasive Method for Induction of
Myocardial Infarction in an Animal Model Using
Tungsten Spirals.” The International Journal of
Cardiovascular Imaging 25 (5): 529–35.
Pleger, S.T., Shan, C., Ksienzyk, J., Bekeredjian, R.,
Boekstegers, P., Hinkel, R., Schinkel, S. et al. 2011.
“Cardiac AAV9-S100A1 Gene Therapy Rescues
Post-Ischemic Heart Failure in a Preclinical Large
Animal Model.” Science Translational Medicine 3
(92): 92ra64.
Poulsen, R.H., Bøtker, H.E. and Rehling, M. 2011.
“Postreperfusion Myocardial Technetium-99mSestamibi Defect Corresponds to Area at Risk:
Experimental Results from an Ischemia-Reperfusion
Porcine Model.” Nuclear Medicine and Biology 38
(6): 819–25.
Prescimone, T., Lionetti, V., Cabiati, M., Caselli,
C., Aquaro, G.D., Matteucci, M., Del Ry, S. and
Giannessi, D. 2013. “Apoptotic Transcriptional
Profile Remains Activated in Late Remodeled Left
Ventricle after Myocardial Infarction in Swine
Infarcted Hearts with Preserved Ejection Fraction.”
References67
Pharmacological Research : The Official Journal of
the Italian Pharmacological Society 70 (1): 41–49.
Size vs Duration of Coronary Occlusion in Dogs.”
Circulation 56 (5): 786–94.
Prescott, M.F., Hasler-Rapacz, J., von Linden-Reed, J.
and Rapacz, J. 1995. “Familial Hypercholesterolemia
Associated with Coronary Atherosclerosis in Swine
Bearing Different Alleles for Apolipoprotein B.”
Annals of the New York Academy of Sciences 748:
283–92; discussion 292–93.
Robich, M.P., Osipov, R.M., Chu, L.M., Han, Y., Feng,
J., Nezafat, R., Clements, R.T., Manning, W.J. and
Sellke, F.W. 2011. “Resveratrol Modifies Risk
Factors for Coronary Artery Disease in Swine with
Metabolic Syndrome and Myocardial Ischemia.”
European Journal of Pharmacology 664 (1-3):
45–53.
Prescott, M.F., McBride, C.H., Hasler-Rapacz, J.,
Von Linden, J. and Rapacz, J. 1991. “Development
of Complex Atherosclerotic Lesions in Pigs with
Inherited Hyper-LDL Cholesterolemia Bearing
Mutant Alleles for Apolipoprotein B.” The American
Journal of Pathology 139 (1): 139–47.
Rodrigo, G.C. and Samani, N.J. 2008. “Ischemic
Preconditioning of the Whole Heart Confers
Protection on Subsequently Isolated Ventricular
Myocytes.” American Journal of Physiology. Heart
and Circulatory Physiology 294 (1): H524–31.
Przyklenk, K., Vivaldi, M.T., Schoen, F.J., Malcolm,
J., Arnold, O. and Kloner, R.A. 1986. “Salvage of
Ischaemic Myocardium by Reperfusion: Importance
of Collateral Blood Flow and Myocardial Oxygen
Demand during Occlusion.” Cardiovascular
Research 20 (6): 403–14.
Rogers, I.S., Nasir, K., Figueroa, A.L., Cury, R.C.,
Hoffmann, U., Vermylen, D.A., Brady, T.J. and
Tawakol, A. 2010. “Feasibility of FDG Imaging of
the Coronary Arteries: Comparison between Acute
Coronary Syndrome and Stable Angina.” JACC.
Cardiovasc.Imaging 3: 388–97.
Pueyo Palazón, C., Alfón, J., Gaffney, P., Berrozpe, M.,
Royo, T. and Badimon, L. 1998. “Effects of Reducing
LDL and Increasing HDL with Gemfibrozil in
Experimental Coronary Lesion Development and
Thrombotic Risk.” Atherosclerosis 136 (2): 333–45.
Rominger, A., Saam, T., Vogl, E., Ubleis, C., la
Fougère, C., Förster, S., Haug, A. et al. 2010.
“In Vivo Imaging of Macrophage Activity in the
Coronary Arteries Using 68Ga-DOTATATE PET/
CT: Correlation with Coronary Calcium Burden
and Risk Factors.” Journal of Nuclear Medicine :
Official Publication, Society of Nuclear Medicine 51
(2): 193–97.
Pätilä, T., Ikonen, T., Kankuri, E., Ahonen, A.,
Krogerus, L., Lauerma, K. and Harjula, A. 2009.
“Spontaneous Recovery of Myocardial Function
after Ligation of Ameroid-Stenosed Coronary
Artery.” Scandinavian Cardiovascular Journal :
SCJ 43 (6): 408–16.
Qu, X., Fang, W., Ye, J., Koh, A.S., Xu, Y., Guan,
S., Li, R. and Shen, Y. 2012. “Acute and Chronic
Myocardial Infarction in a Pig Model: Utility
of Multi-Slice Cardiac Computed Tomography
in Assessing Myocardial Viability and Infarct
Parameters.” European Journal of Radiology 81 (4):
e431–37.
Reffelmann, T., Sensebat, O., Birnbaum, Y., Stroemer,
E., Hanrath, P., Uretsky, B.F. and Schwarz, E.R.
2004. “A Novel Minimal-Invasive Model of Chronic
Myocardial Infarction in Swine.” Coronary Artery
Disease 15 (1): 7–12.
Reimer, K.A. and Jennings, R.B. 1979. “The
‘Wavefront Phenomenon’ of Myocardial Ischemic
Cell Death. II. Transmural Progression of Necrosis
within the Framework of Ischemic Bed Size
(myocardium at Risk) and Collateral Flow.”
Laboratory Investigation; a Journal of Technical
Methods and Pathology 40 (6): 633–44.
Reimer, K.A., Lowe, J.E., Rasmussen, M.M. and
Jennings, R.B. 1977. “The Wavefront Phenomenon
of Ischemic Cell Death. 1. Myocardial Infarct
Roth, D.M., Maruoka, Y., Rogers, J., White,
F.C., Longhurst, J.C. and Bloor, C.M. 1987.
“Development of Coronary Collateral Circulation
in Left Circumflex Ameroid-Occluded Swine
Myocardium.” The American Journal of Physiology
253 (5 Pt 2): H1279–88.
Rud Andersen, H., M ng, M., Thorwest, M. and Falk,
E. 1996. “Remodeling Rather Than Neointimal
Formation Explains Luminal Narrowing After
Deep Vessel Wall Injury : Insights From a Porcine
Coronary (Re)stenosis Model.” Circulation 93 (9):
1716–24.
Rudd, J.H., Warburton, E.A., Fryer, T.D., Jones, H.A.,
Clark, J.C., Antoun, N., Johnstrom, P. et al. 2002.
“Imaging Atherosclerotic Plaque Inflammation
with [18F]-Fluorodeoxyglucose Positron Emission
Tomography.” Circulation 105: 2708–11.
Ruifrok, A.C. and Johnston, D.A. 2001. “Quantification
of Histochemical Staining by Color Deconvolution.”
Anal.Quant.Cytol.Histol. 23: 291–99.
Saeed, M., Hetts, S.W., Do, L., Sullivan, S.M. and
Wilson, M.W. 2013. “MRI Quantification of Left
Ventricular Function in Microinfarct versus Large
Infarct in Swine Model.” The International Journal
of Cardiovascular Imaging 29 (1): 159–68.
68
References
Sahul, Z.H., Mukherjee, R., Song, J., McAteer, J.,
Stroud, R.E., Dione, D.P., Staib, L. et al. 2011.
“Targeted Imaging of the Spatial and Temporal
Variation of Matrix Metalloproteinase Activity
in a Porcine Model of Postinfarct Remodeling:
Relationship
to
Myocardial
Dysfunction.”
Circulation. Cardiovascular Imaging 4 (4): 381–91.
Sakaguchi, G., Sakakibara, Y., Tambara, K., Lu, F.,
Premaratne, G., Nishimura, K. and Komeda, M.
2003. “A Pig Model of Chronic Heart Failure by
Intracoronary Embolization with Gelatin Sponge.”
The Annals of Thoracic Surgery 75 (6): 1942–47.
Sala-Mercado, J.A., Wider, J., Undyala, V.V.R.,
Jahania, S., Yoo, W., Mentzer, R.M., Gottlieb, R.A.
and Przyklenk, K. 2010. “Profound Cardioprotection
with Chloramphenicol Succinate in the Swine
Model of Myocardial Ischemia-Reperfusion Injury.”
Circulation 122 (11 Suppl): S179–84.
Salminen, P.-R., Jonassen, A.K., Aarnes, E.-K., Moen,
C.A., Stangeland, L., Eliassen, F., Kongsvik, R.,
Matre, K., Haaverstad, R. and Grong, K. 2011.
“Antiapoptotic Intervention in Repeated Blood
Cardioplegia: A Porcine Study of Myocardial
Function.” The Annals of Thoracic Surgery 91 (3):
784–91.
Sanz, J., Moreno, P.R. and Fuster, V. 2013. “The Year
in Atherothrombosis.” Journal of the American
College of Cardiology 62 (13): 1131–43.
Saraste, A., Kajander, S., Han, C., Nesterov, S. V
and Knuuti, J. 2012. “PET: Is Myocardial Flow
Quantification a Clinical Reality?” Journal of
Nuclear Cardiology : Official Publication of the
American Society of Nuclear Cardiology 19 (5):
1044–59.
Sassen, L.M., den Boer, M.O., Rensen, R.J.,
Saxena, P.R. and Verdouw, P.D. 1988. “Bisoprolol
Improves Perfusion of Ischaemic Myocardium
in Anaesthetized Pigs.” British Journal of
Pharmacology 95 (2): 361–70.
Schelbert, H.R., Wisenberg, G., Phelps, M.E., Gould,
K.L., Henze, E., Hoffman, E.J., Gomes, A. and
Kuhl, D.E. 1982. “Noninvasive Assessment of
Coronary Stenoses by Myocardial Imaging during
Pharmacologic Coronary Vasodilation. VI. Detection
of Coronary Artery Disease in Human Beings with
Intravenous N-13 Ammonia and Positron Computed
Tomography.” The American Journal of Cardiology
49 (5): 1197–1207.
Schneider, C., Jaquet, K., Geidel, S., Malisius, R.,
Boczor, S., Rau, T., Zienkiewicz, T., Hennig, D.,
Kuck, K.-H. and Krause, K. 2010. “Regional
Diastolic and Systolic Function by Strain Rate
Imaging for the Detection of Intramural Viability
during Dobutamine Stress Echocardiography
in a Porcine Model of Myocardial Infarction.”
Echocardiography 27 (5): 552–62.
Scholtz, L., Sarkin, A. and Lockhat, Z. 2014. “Current
Clinical Applications of Cardiovascular Magnetic
Resonance Imaging.” Cardiovascular Journal of
Africa 25 (4): 185–90.
Schuleri, K.H., Boyle, A.J., Centola, M., Amado,
L.C., Evers, R., Zimmet, J.M., Evers, K.S. et al.
2008. “The Adult Göttingen Minipig as a Model for
Chronic Heart Failure after Myocardial Infarction:
Focus on Cardiovascular Imaging and Regenerative
Therapies.” Comparative Medicine 58 (6): 568–79.
Schwartz, R.S., Huber, K.C., Murphy, J.G., Edwards,
W.D., Camrud, A.R., Vlietstra, R.E. and Holmes,
D.R. 1992. “Restenosis and the Proportional
Neointimal Response to Coronary Artery Injury:
Results in a Porcine Model.” Journal of the
American College of Cardiology 19 (2): 267–74.
Schwartz, R.S., Murphy, J.G., Edwards, W.D., Camrud,
A.R., Vliestra, R.E. and Holmes, D.R. 1990.
“Restenosis after Balloon Angioplasty. A Practical
Proliferative Model in Porcine Coronary Arteries.”
Circulation 82 (6): 2190–2200.
Seol, S.-H. and Lindner, J.R. 2014. “A Primer
on the Methods and Applications for Contrast
Echocardiography in Clinical Imaging.” Journal of
Cardiovascular Ultrasound 22 (3): 101–10.
Sheriff, A., Schindler, R., Vogt, B., Abdel-Aty, H.,
Unger, J.K., Bock, C., Gebauer, F. et al. 2014.
“Selective Apheresis of C-Reactive Protein: A New
Therapeutic Option in Myocardial Infarction?”
Journal of Clinical Apheresis 30(1):15-21.
Shimokawa, H., Tomoike, H., Nabeyama, S.,
Yamamoto, H., Ishii, Y., Tanaka, K. and Nakamura,
M. 1985. “Coronary Artery Spasm Induced in
Miniature Swine: Angiographic Evidence and
Relation to Coronary Atherosclerosis.” American
Heart Journal 110 (2): 300–310.
Shimokawa, H. and Vanhoutte, P.M. 1988.
“Dietary Cod-Liver Oil Improves EndotheliumDependent Responses in Hypercholesterolemic
and Atherosclerotic Porcine Coronary Arteries.”
Circulation 78 (6): 1421–30.
Shinohara, G., Morita, K., Nagahori, R., Koh, Y.,
Kinouchi, K., Abe, T. and Hashimoto, K. 2011.
“Ischemic
Postconditioning
Promotes
Left
Ventricular Functional Recovery after Cardioplegic
Arrest in an in Vivo Piglet Model of Global Ischemia
Reperfusion Injury on Cardiopulmonary Bypass.”
The Journal of Thoracic and Cardiovascular
Surgery 142 (4): 926–32.
Shudo, Y., Miyagawa, S., Fukushima, S., Saito, A.,
Shimizu, T., Okano, T. and Sawa, Y. 2011. “Novel
References69
Regenerative Therapy Using Cell-Sheet Covered
with Omentum Flap Delivers a Huge Number of
Cells in a Porcine Myocardial Infarction Model.”
The Journal of Thoracic and Cardiovascular
Surgery 142 (5): 1188–96.
Shuros, A.C., Salo, R.W., Florea, V.G., Pastore, J.,
Kuskowski, M.A., Chandrashekhar, Y. and Anand,
I.S. 2007. “Ventricular Preexcitation Modulates
Strain and Attenuates Cardiac Remodeling in a
Swine Model of Myocardial Infarction.” Circulation
116 (10): 1162–69.
Silva, G. V, Fernandes, M.R., Madonna, R., Clubb,
F., Oliveira, E., Jimenez-Quevedo, P., Branco, R.
et al. 2009. “Comparative Healing Response after
Sirolimus- and Paclitaxel-Eluting Stent Implantation
in a Pig Model of Restenosis.” Catheterization and
Cardiovascular Interventions : Official Journal of
the Society for Cardiac Angiography & Interventions
73 (6): 801–8.
Silvola, J.M.U., Saraste, A., Forsback, S., Laine,
V.J.O., Saukko, P., Heinonen, S.E., Ylä-Herttuala,
S., Roivainen, A. and Knuuti, J. 2011. “Detection of
Hypoxia by [18F]EF5 in Atherosclerotic Plaques in
Mice.” Arteriosclerosis, Thrombosis, and Vascular
Biology 31 (5): 1011–15.
Skyschally, A., Walter, B., Schultz Hansen, R. and
Heusch, G. 2013. “The Antiarrhythmic Dipeptide
ZP1609 (danegaptide) When given at Reperfusion
Reduces Myocardial Infarct Size in Pigs.” NaunynSchmiedeberg’s Archives of Pharmacology 386 (5):
383–91.
Skålén, K., Gustafsson, M., Rydberg, E.K., Hultén,
L.M., Wiklund, O., Innerarity, T.L. and Borén, J.
2002. “Subendothelial Retention of Atherogenic
Lipoproteins in Early Atherosclerosis.” Nature 417
(6890): 750–54.
So, A., Hsieh, J., Li, J.-Y., Hadway, J., Kong, H.-F. and
Lee, T.-Y. 2012. “Quantitative Myocardial Perfusion
Measurement Using CT Perfusion: A Validation
Study in a Porcine Model of Reperfused Acute
Myocardial Infarction.” The International Journal
of Cardiovascular Imaging 28 (5): 1237–48.
Sodha, N.R., Clements, R.T., Feng, J., Liu, Y., Bianchi,
C., Horvath, E.M., Szabo, C., Stahl, G.L. and Sellke,
F.W. 2009. “Hydrogen Sulfide Therapy Attenuates
the Inflammatory Response in a Porcine Model
of Myocardial Ischemia/reperfusion Injury.” The
Journal of Thoracic and Cardiovascular Surgery
138 (4): 977–84.
Sorop, O., Merkus, D., de Beer, V.J., Houweling, B.,
Pistea, A., McFalls, E.O., Boomsma, F. et al. 2008.
“Functional and Structural Adaptations of Coronary
Microvessels Distal to a Chronic Coronary Artery
Stenosis.” Circulation Research 102 (7): 795–803.
St. Louis, J.D., Hughes, G.C., Kypson, A.P., DeGrado,
T.R., Donovan, C.L., Coleman, R.E., Yin, B.,
Steenbergen, C., Landolfo, K.P. and Lowe, J.E. 2000.
“An Experimental Model of Chronic Myocardial
Hibernation.” The Annals of Thoracic Surgery 69
(5): 1351–57.
Stanley, W.C. 2001. “Cardiac Energetics during
Ischaemia and the Rationale for Metabolic
Interventions.” Coronary Artery Disease 12 Suppl
1: S3–7.
Stanley, W.C. 2004. “Myocardial Energy Metabolism
during Ischemia and the Mechanisms of
Metabolic Therapies.” Journal of Cardiovascular
Pharmacology and Therapeutics 9 Suppl 1
(September): S31–45.
Stary, H.C. 2000. “Lipid and Macrophage
Accumulations in Arteries of Children and the
Development of Atherosclerosis.” The American
Journal of Clinical Nutrition 72 (5 Suppl): 1297S
– 1306S.
Swindle, M.M., Makin, A., Herron, A.J., Clubb,
F.J. and Frazier, K.S. 2012. “Swine as Models in
Biomedical Research and Toxicology Testing.”
Veterinary Pathology 49 (2): 344–56.
Swynghedauw, B. 1999. “Molecular Mechanisms of
Myocardial Remodeling.” Physiol Rev. 79: 215–62.
Szilárd, M., Mesotten, L., Maes, A., Liu, X., Nuyts,
J., Bormans, G., De Groot, T. et al. 2000. “A
Nonsurgical Porcine Model of Left Ventricular
Dysfunction. Validation of Myocardial Viability
Using Dobutamine Stress Echocardiography and
Positron Emission Tomography.” International
Journal of Cardiovascular Interventions 3 (2):
111–20.
Tahara, N., Mukherjee, J., de Haas, H.J., Petrov, A.D.,
Tawakol, A., Haider, N., Tahara, A. et al. 2014.
“2-Deoxy-2-[18F]fluoro-D-Mannose
Positron
Emission Tomography Imaging in Atherosclerosis.”
Nature Medicine 20 (2): 215–19.
Tanaka, Y., Genet, M., Lee, L.C., Martin, A.J.,
Sievers, R., Gerstenfeld, E.P. and Chuan Lee, L.
2014. “Utility of High-Resolution Electroanatomic
Mapping of the Left Ventricle Using a Multispline
Basket Catheter in a Swine Model of Chronic
Myocardial Infarction.” Heart Rhythm : The Official
Journal of the Heart Rhythm Society, August.
Tarkin, J.M., Joshi, F.R. and Rudd, J.H.F. 2014. “PET
Imaging of Inflammation in Atherosclerosis.”
Nature Reviews. Cardiology 11 (8): 443–57.
Tellez, A., Krueger, C.G., Seifert, P., Winsor-Hines,
D., Piedrahita, C., Cheng, Y., Milewski, K. et al.
2010. “Coronary Bare Metal Stent Implantation
in Homozygous LDL Receptor Deficient Swine
70
References
Induces a Neointimal Formation Pattern Similar to
Humans.” Atherosclerosis 213 (2): 518–24.
Tellez, A., Schuster, D.S., Alviar, C., López-Berenstein,
G., Sanguino, A., Ballantyne, C., Perrard, X.-Y.D.
et al. 2011. “Intramural Coronary Lipid Injection
Induces Atheromatous
Lesions
Expressing
Proinflammatory
Chemokines:
Implications
for the Development of a Porcine Model of
Atherosclerosis.” Cardiovascular Revascularization
Medicine : Including Molecular Interventions 12
(5): 304–11.
Teramoto, N., Koshino, K., Yokoyama, I., Miyagawa,
S., Zeniya, T., Hirano, Y., Fukuda, H. et al. 2011.
“Experimental Pig Model of Old Myocardial
Infarction with Long Survival Leading to Chronic
Left Ventricular Dysfunction and Remodeling as
Evaluated by PET.” J.Nucl.Med. 52: 761–68.
Thim, T., Hagensen, M.K., Drouet, L., Bal Dit Sollier,
C., Bonneau, M., Granada, J.F., Nielsen, L.B.,
Paaske, W.P., Bøtker, H.E. and Falk, E. 2010.
“Familial Hypercholesterolaemic Downsized Pig
with Human-like Coronary Atherosclerosis: A
Model for Preclinical Studies.” EuroIntervention :
Journal of EuroPCR in Collaboration with the
Working Group on Interventional Cardiology of the
European Society of Cardiology 6 (2): 261–68.
Thim, T., Hagensen, M.K., Wallace-Bradley, D.,
Granada, J.F., Kaluza, G.L., Drouet, L., Paaske,
W.P., Bøtker, H.E. and Falk, E. 2010. “Unreliable
Assessment of Necrotic Core by Virtual Histology
Intravascular Ultrasound in Porcine Coronary Artery
Disease.” Circulation. Cardiovascular Imaging 3
(4): 384–91.
Thomas, S.A., Fallavollita, J.A., Lee, T.-C., Feng,
J. and Canty, J.M. 1999. “Absence of Troponin I
Degradation or Altered Sarcoplasmic Reticulum
Uptake Protein Expression After Reversible
Ischemia in Swine.” Circulation Research 85 (5):
446–56.
Thorpe, P.E., Hunter, W.J., Dovgan, P.S. and Agrawal,
D.K. 1996. “A Noninjury, Diet-Induced Swine
Model of Atherosclerosis for CardiovascularInterventional Research.” Angiology 47 (9): 849–58.
Tillisch, J., Brunken, R., Marshall, R., Schwaiger,
M., Mandelkern, M., Phelps, M. and Schelbert,
H. 1986. “Reversibility of Cardiac Wall-Motion
Abnormalities Predicted by Positron Tomography.”
New England Journal of Medicine 314 (14): 884–88.
Torrado, M., Iglesias, R., Centeno, A., López, E. and
Mikhailov, A.T. 2011. “Targeted Gene-Silencing
Reveals the Functional Significance of Myocardin
Signaling in the Failing Heart.” PloS One 6 (10):
e26392.
Turk, J.R., Henderson, K.K., Vanvickle, G.D., Watkins,
J. and Laughlin, M.H. 2005. “Arterial Endothelial
Function in a Porcine Model of Early Stage
Atherosclerotic Vascular Disease.” International
Journal of Experimental Pathology 86 (5): 335–45.
Tuunanen, H. and Knuuti, J. 2011. “Metabolic
Remodelling
in
Human
Heart
Failure.”
Cardiovascular Research 90 (2): 251–57.
Tuzun, E., Oliveira, E., Narin, C., Khalil, H.,
Jimenez-Quevedo, P., Perin, E. and Silva, G. 2010.
“Correlation of Ischemic Area and Coronary Flow
with Ameroid Size in a Porcine Model.” The Journal
of Surgical Research 164 (1): 38–42.
Ukkonen, H., Saraste, M., Akkila, J., Knuuti, J.,
Karanko, M., Iida, H., Lehikoinen, P., Nagren,
K., Lehtonen, L. and Voipio-Pulkki, L.M. 2000.
“Myocardial Efficiency during Levosimendan
Infusion in Congestive Heart Failure.” Clin.
Pharmacol.Ther. 68: 522–31.
Ukkonen, H., Tops, L., Saraste, A., Naum, A.,
Koistinen, J., Bax, J. and Knuuti, J. 2009. “The
Effect of Right Ventricular Pacing on Myocardial
Oxidative Metabolism and Efficiency: Relation with
Left Ventricular Dyssynchrony.” Eur.J.Nucl.Med.
Mol.Imaging 36: 2042–48.
Waksman, R., Fournadjiev, J., Baffour, R., Pakala,
R., Hellinga, D., Leborgne, L., Yazdi, H. et al.
2004. “Transepicardial Autologous Bone MarrowDerived Mononuclear Cell Therapy in a Porcine
Model of Chronically Infarcted Myocardium.”
Cardiovascular Radiation Medicine 5 (3): 125–31.
Van den Hoff, J., Burchert, W., Borner, A.R., Fricke,
H., Kuhnel, G., Meyer, G.J., Otto, D., Weckesser,
E., Wolpers, H.G. and Knapp, W.H. 2001. “[1-(11)
C]Acetate as a Quantitative Perfusion Tracer in
Myocardial PET.” J.Nucl.Med. 42: 1174–82.
Van der Pals, J., Koul, S., Andersson, P., Götberg,
M., Ubachs, J.F.A., Kanski, M., Arheden, H.,
Olivecrona, G.K., Larsson, B. and Erlinge, D.
2010. “Treatment with the C5a Receptor Antagonist
ADC-1004 Reduces Myocardial Infarction in
a Porcine Ischemia-Reperfusion Model.” BMC
Cardiovascular Disorders 10: 45.
Van Hout, G.P.J., de Jong, R., Vrijenhoek, J.E.P.,
Timmers, L., Duckers, H.J. and Hoefer, I.E.
2013. “Admittance-Based Pressure-Volume Loop
Measurements in a Porcine Model of Chronic
Myocardial Infarction.” Experimental Physiology
98 (11): 1565–75.
Vanoverschelde, J.L., Wijns, W., Depré, C., Essamri,
B., Heyndrickx, G.R., Borgers, M., Bol, A. and
Melin, J.A. 1993. “Mechanisms of Chronic
Regional Postischemic Dysfunction in Humans.
References71
New Insights from the Study of Noninfarcted
Collateral-Dependent Myocardium.” Circulation 87
(5): 1513–23.
Varga-Szemes, A., Simor, T., Lenkey, Z., van der
Geest, R.J., Kirschner, R., Toth, L., Brott, B.C.,
Elgavish, A. and Elgavish, G.A. 2014. “Infarct
Density Distribution by MRI in the Porcine Model
of Acute and Chronic Myocardial Infarction as a
Potential Method Transferable to the Clinic.” The
International Journal of Cardiovascular Imaging 30
(5): 937–48.
Watanabe, I., Johnson, T.A., Buchanan, J., Engle, C.L.
and Gettes, L.S. 1987. “Effect of Graded Coronary
Flow Reduction on Ionic, Electrical, and Mechanical
Indexes of Ischemia in the Pig.” Circulation 76 (5):
1127–34.
Weaver, M.E., Pantely, G.A., Bristow, J.D. and Ladley,
H.D. 1986. “A Quantitative Study of the Anatomy
and Distribution of Coronary Arteries in Swine
in Comparison with Other Animals and Man.”
Cardiovascular Research 20 (12): 907–17.
Weiss, S.M. and Saint, D.A. 2010. “The Persistent
Sodium Current Blocker Riluzole Is Antiarrhythmic
and Anti-Ischaemic in a Pig Model of Acute
Myocardial Infarction.” PloS One 5 (11): e14103.
Wheeler, D.G., Joseph, M.E., Mahamud, S.D., Aurand,
W.L., Mohler, P.J., Pompili, V.J., Dwyer, K.M. et
al. 2012. “Transgenic Swine: Expression of Human
CD39 Protects against Myocardial Injury.” Journal
of Molecular and Cellular Cardiology 52 (5): 958–
61.
Wiggers, H., Klebe, T., Heickendorff, L., Høst, N.B.,
Danielsen, C.C., Baandrup, U. and Andersen, H.R.
1997. “Ischemia and Reperfusion of the Porcine
Myocardium: Effect on Collagen.” Journal of
Molecular and Cellular Cardiology 29 (1): 289–99.
Vilahur, G., Casani, L., Peña, E., Juan-Babot, O.,
Mendieta, G., Crespo, J. and Badimon, L. 2014.
“HMG-CoA Reductase Inhibition Prior Reperfusion
Improves Reparative Fibrosis Post-Myocardial
Infarction in a Preclinical Experimental Model.”
International Journal of Cardiology 175 (3): 528–
38.
Wilensky, R.L., Shi, Y., Mohler, E.R., Hamamdzic, D.,
Burgert, M.E., Li, J., Postle, A. et al. 2008. “Inhibition
of Lipoprotein-Associated Phospholipase A2
Reduces Complex Coronary Atherosclerotic Plaque
Development.” Nature Medicine 14 (10): 1059–66.
Virmani, R., Kolodgie, F.D., Burke, A.P., Farb, A.
and Schwartz, S.M. 2000. “Lessons from Sudden
Coronary Death: A Comprehensive Morphological
Classification Scheme for Atherosclerotic Lesions.”
Arteriosclerosis, Thrombosis, and Vascular Biology
20 (5): 1262–75.
Wojakowski, W., Tendera, M., Cybulski, W., ZubaSurma, E.K., Szade, K., Florczyk, U., Kozakowska,
M. et al. 2012. “Effects of Intracoronary Delivery
of Allogenic Bone Marrow-Derived Stem Cells
Expressing Heme Oxygenase-1 on Myocardial
Reperfusion Injury.” Thrombosis and Haemostasis
108 (3): 464–75.
Wolpers, H.G., Buck, A., Nguyen, N., Marcowitz,
P.A., Armstrong, W.F., Starling, M.R., Hicks,
R., Mangner, T.J. and Schwaiger, M. 1994. “An
Approach to Ventricular Efficiency by Use of
Carbon 11-Labeled Acetate and Positron Emission
Tomography.” Journal of Nuclear Cardiology :
Official Publication of the American Society of
Nuclear Cardiology 1 (3): 262–69.
Wolpers, H.G., Burchert, W., van den Hoff, J.,
Weinhardt, R., Meyer, G.J. and Lichtlen, P.R. 1997.
“Assessment of Myocardial Viability by Use of
11C-Acetate and Positron Emission Tomography.
Threshold Criteria of Reversible Dysfunction.”
Circulation 95 (0009-7322 (Print)). Department
of Internal Medicine, Hannover Medical School,
Germany: 1417–24.
Von Degenfeld, G., Raake, P., Kupatt, C., Lebherz,
C., Hinkel, R., Gildehaus, F.J., Münzing, W. et al.
2003. “Selective Pressure-Regulated Retroinfusion
of Fibroblast Growth Factor-2 into the Coronary
Vein Enhances Regional Myocardial Blood Flow
and Function in Pigs with Chronic Myocardial
Ischemia.” Journal of the American College of
Cardiology 42 (6): 1120–28.
Worthley, S.G., Helft, G., Fuster, V., Fayad, Z.A.,
Fallon, J.T., Osende, J.I., Roqué, M. et al. 2000. “High
Resolution Ex Vivo Magnetic Resonance Imaging
of in Situ Coronary and Aortic Atherosclerotic
Plaque in a Porcine Model.” Atherosclerosis 150
(2): 321–29.
Worthley, S.G., Helft, G., Fuster, V., Fayad, Z.A.,
Rodriguez, O.J., Zaman, A.G., Fallon, J.T. and
Badimon, J.J. 2000. “Noninvasive In Vivo Magnetic
Resonance Imaging of Experimental Coronary
Artery Lesions in a Porcine Model.” Circulation 101
(25): 2956–61.
Wu, M., Bogaert, J., D’hooge, J., Sipido, K., Maes,
F., Dymarkowski, S., Rademakers, F.E. and Claus,
P. 2010. “Closed-Chest Animal Model of Chronic
Coronary Artery Stenosis. Assessment with
Magnetic Resonance Imaging.” The International
Journal of Cardiovascular Imaging 26 (3): 299–308.
Wu, M., D’hooge, J., Ganame, J., Ferferieva, V.,
Sipido, K.R., Maes, F., Dymarkowski, S., Bogaert,
J., Rademakers, F.E. and Claus, P. 2011. “Non-
72
References
Invasive Characterization of the Area-at-Risk
Using Magnetic Resonance Imaging in Chronic
Ischaemia.” Cardiovascular Research 89 (1): 166–
74.
Wykrzykowska, J., Lehman, S., Williams, G.,
Parker, J.A., Palmer, M.R., Varkey, S., Kolodny,
G. and Laham, R. 2009. “Imaging of Inflamed
and Vulnerable Plaque in Coronary Arteries with
18F-FDG PET/CT in Patients with Suppression of
Myocardial Uptake Using a Low-Carbohydrate,
High-Fat Preparation.” J.Nucl.Med. 50: 563–68.
Vähäsilta, T., Saraste, A., Kytö, V., Malmberg, M.,
Kiss, J., Kentala, E., Kallajoki, M. and Savunen, T.
2005. “Cardiomyocyte Apoptosis after Antegrade
and Retrograde Cardioplegia.” The Annals of
Thoracic Surgery 80 (6): 2229–34.
Yan, Z. and Hansson, G.K. 2007. “Innate Immunity,
Macrophage Activation, and Atherosclerosis.”
Immunological Reviews 219: 187–203.
Yang, K., Xiang, P., Zhang, C., Zou, L., Wu, X., Gao,
Y., Kang, Z., He, K., Liu, J. and Peng, C. 2011.
“Magnetic Resonance Evaluation of Transplanted
Mesenchymal Stem Cells after Myocardial
Infarction in Swine.” The Canadian Journal of
Cardiology 27 (6): 818–25.
Zhang, J., Wilke, N., Wang, Y., Zhang, Y., Wang,
C., Eijgelshoven, M.H., Cho, Y.K. et al. 1996.
“Functional and Bioenergetic Consequences of
Postinfarction Left Ventricular Remodeling in a
New Porcine Model. MRI and 31 P-MRS Study.”
Circulation 94: 1089–1100.
Zhang, L., Zalewski, A., Liu, Y., Mazurek, T., Cowan,
S., Martin, J.L., Hofmann, S.M., Vlassara, H. and
Shi, Y. 2003. “Diabetes-Induced Oxidative Stress
and Low-Grade Inflammation in Porcine Coronary
Arteries.” Circulation 108 (4): 472–78.
Zhu, X.-Y., Zhang, Z.-L., Li, P., Liang, W.-Y., Feng,
X.-R. and Liu, M.-L. 2013. “Shenyuan, an Extract
of American Ginseng and Corydalis Tuber Formula,
Attenuates Cardiomyocyte Apoptosis via Inhibition
of Endoplasmic Reticulum Stress and Oxidative
Stress in a Porcine Model of Acute Myocardial
Infarction.” Journal of Ethnopharmacology 150 (2):
672–81.